Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 292
Filtrar
1.
Mol Cell ; 81(24): 5039-5051.e5, 2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34784509

RESUMO

Cyclic oligonucleotide-based antiphage signaling systems (CBASS) are antiviral defense operons that protect bacteria from phage replication. Here, we discover a widespread class of CBASS transmembrane (TM) effector proteins that respond to antiviral nucleotide signals and limit phage propagation through direct membrane disruption. Crystal structures of the Yersinia TM effector Cap15 reveal a compact 8-stranded ß-barrel scaffold that forms a cyclic dinucleotide receptor domain that oligomerizes upon activation. We demonstrate that activated Cap15 relocalizes throughout the cell and specifically induces rupture of the inner membrane. Screening for active effectors, we identify the function of distinct families of CBASS TM effectors and demonstrate that cell death via disruption of inner-membrane integrity is a common mechanism of defense. Our results reveal the function of the most prominent class of effector protein in CBASS immunity and define disruption of the inner membrane as a widespread strategy of abortive infection in bacterial phage defense.


Assuntos
Proteínas de Bactérias/metabolismo , Bacteriófagos/patogenicidade , Membrana Celular/virologia , Escherichia coli/virologia , Yersinia/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bacteriófagos/imunologia , Morte Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli/genética , Escherichia coli/imunologia , Escherichia coli/metabolismo , Interações Hospedeiro-Patógeno , Ligantes , Conformação Proteica , Multimerização Proteica , Transporte Proteico , Transdução de Sinais , Relação Estrutura-Atividade , Yersinia/genética
2.
J Biol Chem ; 300(6): 107331, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38703997

RESUMO

Mono-O-glycosylation of target proteins by bacterial toxins or effector proteins is a well-known mechanism by which bacteria interfere with essential functions of host cells. The respective glycosyltransferases are important virulence factors such as the Clostridioides difficile toxins A and B. Here, we describe two glycosyltransferases of Yersinia species that have a high sequence identity: YeGT from the zoonotic pathogen Yersinia enterocolitica and YkGT from the murine pathogen Yersinia kristensenii. We show that both modify Rho family proteins by attachment of GlcNAc at tyrosine residues (Tyr-34 in RhoA). Notably, the enzymes differed in their target protein specificity. While YeGT modified RhoA, B, and C, YkGT possessed a broader substrate spectrum and glycosylated not only Rho but also Rac and Cdc42 subfamily proteins. Mutagenesis studies indicated that residue 177 is important for this broader target spectrum. We determined the crystal structure of YeGT shortened by 16 residues N terminally (sYeGT) in the ligand-free state and bound to UDP, the product of substrate hydrolysis. The structure assigns sYeGT to the GT-A family. It shares high structural similarity to glycosyltransferase domains from toxins. We also demonstrated that the 16 most N-terminal residues of YeGT and YkGT are important for the mediated translocation into the host cell using the pore-forming protective antigen of anthrax toxin. Mediated introduction into HeLa cells or ectopic expression of YeGT and YkGT caused morphological changes and redistribution of the actin cytoskeleton. The data suggest that YeGT and YkGT are likely bacterial effectors belonging to the family of tyrosine glycosylating bacterial glycosyltransferases.


Assuntos
Proteínas de Bactérias , Tirosina , Yersinia , Glicosilação , Humanos , Yersinia/metabolismo , Yersinia/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Tirosina/metabolismo , Tirosina/química , Glicosiltransferases/metabolismo , Glicosiltransferases/genética , Glicosiltransferases/química , Proteína rhoA de Ligação ao GTP/metabolismo , Yersinia enterocolitica/metabolismo , Yersinia enterocolitica/genética , Animais , Células HeLa , Camundongos , Cristalografia por Raios X , Yersiniose/metabolismo , Yersiniose/microbiologia
3.
Annu Rev Microbiol ; 74: 221-245, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32660389

RESUMO

Microbial pathogens have evolved complex mechanisms to interface with host cells in order to evade host defenses and replicate. However, mammalian innate immune receptors detect the presence of molecules unique to the microbial world or sense the activity of virulence factors, activating antimicrobial and inflammatory pathways. We focus on how studies of the major virulence factor of one group of microbial pathogens, the type III secretion system (T3SS) of human pathogenic Yersinia, have shed light on these important innate immune responses. Yersinia are largely extracellular pathogens, yet they insert T3SS cargo into target host cells that modulate the activity of cytosolic innate immune receptors. This review covers both the host pathways that detect the Yersinia T3SS and the effector proteins used by Yersinia to manipulate innate immune signaling.


Assuntos
Citosol/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Sistemas de Secreção Tipo III/imunologia , Yersinia/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Citosol/microbiologia , Humanos , Inflamassomos , Piroptose , Transdução de Sinais , Fatores de Virulência/metabolismo , Yersinia/metabolismo , Yersinia/patogenicidade
4.
PLoS Genet ; 18(7): e1010321, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35901167

RESUMO

The type III secretion system (T3SS) is an appendage used by many bacterial pathogens, such as pathogenic Yersinia, to subvert host defenses. However, because the T3SS is energetically costly and immunogenic, it must be tightly regulated in response to environmental cues to enable survival in the host. Here we show that expression of the Yersinia Ysc T3SS master regulator, LcrF, is orchestrated by the opposing activities of the repressive H-NS/YmoA histone-like protein complex and induction by the iron and oxygen-regulated IscR transcription factor. While deletion of iscR or ymoA has been shown to decrease and increase LcrF expression and type III secretion, respectively, the role of H-NS in this system has not been definitively established because hns is an essential gene in Yersinia. Using CRISPRi knockdown of hns, we show that hns depletion causes derepression of lcrF. Furthermore, we find that while YmoA is dispensable for H-NS binding to the lcrF promoter, YmoA binding to H-NS is important for H-NS repressive activity. We bioinformatically identified three H-NS binding regions within the lcrF promoter and demonstrate binding of H-NS to these sites in vivo using chromatin immunoprecipitation. Using promoter truncation and binding site mutation analysis, we show that two of these H-NS binding regions are important for H-NS/YmoA-mediated repression of the lcrF promoter. Surprisingly, we find that IscR is dispensable for lcrF transcription in the absence of H-NS/YmoA. Indeed, IscR-dependent regulation of LcrF and type III secretion in response to changes in oxygen, such as those Yersinia is predicted to experience during host infection, only occurs in the presence of an H-NS/YmoA complex. These data suggest that, in the presence of host tissue cues that drive sufficient IscR expression, IscR can act as a roadblock to H-NS/YmoA-dependent repression of RNA polymerase at the lcrF promoter to turn on T3SS expression.


Assuntos
Regulação Bacteriana da Expressão Gênica , Yersinia , Proteínas de Bactérias/metabolismo , Histonas/genética , Oxigênio/metabolismo , Yersinia/genética , Yersinia/metabolismo
5.
PLoS Pathog ; 18(5): e1010251, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35604950

RESUMO

Yersinia enterocolitica employs a type three secretion system (T3SS) to translocate immunosuppressive effector proteins into host cells. To this end, the T3SS assembles a translocon/pore complex composed of the translocator proteins YopB and YopD in host cell membranes serving as an entry port for the effectors. The translocon is formed in a Yersinia-containing pre-phagosomal compartment that is connected to the extracellular space. As the phagosome matures, the translocon and the membrane damage it causes are recognized by the cell-autonomous immune system. We infected cells in the presence of fluorophore-labeled ALFA-tag-binding nanobodies with a Y. enterocolitica strain expressing YopD labeled with an ALFA-tag. Thereby we could record the integration of YopD into translocons and its intracellular fate in living host cells. YopD was integrated into translocons around 2 min after uptake of the bacteria into a phosphatidylinositol-4,5-bisphosphate enriched pre-phagosomal compartment and remained there for 27 min on average. Damaging of the phagosomal membrane as visualized with recruitment of GFP-tagged galectin-3 occurred in the mean around 14 min after translocon formation. Shortly after recruitment of galectin-3, guanylate-binding protein 1 (GBP-1) was recruited to phagosomes, which was accompanied by a decrease in the signal intensity of translocons, suggesting their degradation or disassembly. In sum, we were able for the first time to film the spatiotemporal dynamics of Yersinia T3SS translocon formation and degradation and its sensing by components of the cell-autonomous immune system.


Assuntos
Yersinia pseudotuberculosis , Yersinia , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Galectina 3 , Sistemas de Secreção Tipo III/metabolismo , Yersinia/metabolismo , Yersinia pseudotuberculosis/metabolismo
6.
Appl Environ Microbiol ; 89(7): e0024023, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37338394

RESUMO

Metal ions are essential nutrients for all life forms, and restriction of metal ion availability is an effective host defense against bacterial infection. Meanwhile, bacterial pathogens have developed equally effective means to secure their metal ion supply. The enteric pathogen Yersinia pseudotuberculosis was found to uptake zinc using the T6SS4 effector YezP, which is essential for Zn2+ acquisition and bacterial survival under oxidative stress. However, the mechanism of this zinc uptake pathway has not been fully elucidated. Here, we identified the hemin uptake receptor HmuR for YezP, which can mediate import of Zn2+ into the periplasm by the YezP-Zn2+ complex and demonstrated that YezP functions extracellularly. This study also confirmed that the ZnuCB transporter is the inner membrane transporter for Zn2+ from the periplasm to cytoplasm. Overall, our results reveal the complete T6SS/YezP/HmuR/ZnuABC pathway, wherein multiple systems are coupled to support zinc uptake by Y. pseudotuberculosis under oxidative stress. IMPORTANCE Identifying the transporters involved in import of metal ions under normal physiological growth conditions in bacterial pathogens will clarify its pathogenic mechanism. Y. pseudotuberculosis YPIII, a common foodborne pathogen that infects animals and humans, uptake zinc via the T6SS4 effector YezP. However, the outer and inner transports involved in Zn2+ acquisition remain unknown. The important outcomes of this study are the identification of the hemin uptake receptor HmuR and inner membrane transporter ZnuCB that import Zn2+ into the cytoplasm via the YezP-Zn2+ complex, and elucidation of the complete Zn2+ acquisition pathway consisting of T6SS, HmuRSTUV, and ZnuABC, thereby providing a comprehensive view of T6SS-mediated ion transport and its functions.


Assuntos
Hemina , Infecções por Yersinia pseudotuberculosis , Humanos , Animais , Hemina/metabolismo , Yersinia/metabolismo , Transporte Biológico , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Zinco/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
7.
Molecules ; 27(9)2022 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-35566248

RESUMO

A high enzyme-yield strain Yersinia sp. 298 was screened from marine bacteria harvested from the coastal water. The screening conditions were extensive, utilizing hyaluronic acid (HA)/chondroitin sulfate (CS) as the carbon source. A coding gene yshyl8A of the family 8 polysaccharide lyase (PL8) was cloned from the genome of Yersinia sp. 298 and subjected to recombinant expression. The specific activity of the recombinase YsHyl8A was 11.19 U/mg, with an optimal reaction temperature of 40 °C and 50% of its specific activity remaining after thermal incubation at 30 °C for 1 h. In addition, its optimal reaction pH was 7.5, and while it was most stable at pH 6.0 in Na2HPO4-citric acid buffer, it remained highly stable at pH 6.0-11.0. Further, its enzymatic activity was increased five-fold with 0.1 M NaCl. YsHyl8A, as an endo-lyase, can degrade both HA and CS, producing disaccharide end-products. These properties suggested that YsHyl8A possessed both significant alkalophilic and cold-adapted features while being dependent on NaCl, likely resulting from its marine source. Yersinia is a typical fish pathogen, with glycosaminoglycan lyase (GAG lyase) as a potential pathogenic factor, exhibiting strong hyaluronidase and chondroitinase activity. Further research on the pathogenic mechanism of GAG lyase may benefit the prevention and treatment of related diseases.


Assuntos
Glicosaminoglicanos , Liases , Animais , Sulfatos de Condroitina , Ácido Hialurônico/química , Concentração de Íons de Hidrogênio , Polissacarídeo-Liases/química , Cloreto de Sódio , Yersinia/genética , Yersinia/metabolismo
8.
PLoS Pathog ; 15(12): e1008001, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31869388

RESUMO

The enteropathogen Yersinia pseudotuberculosis and the related plague agent Y. pestis require the Ysc type III secretion system (T3SS) to subvert phagocyte defense mechanisms and cause disease. Yet type III secretion (T3S) in Yersinia induces growth arrest and innate immune recognition, necessitating tight regulation of the T3SS. Here we show that Y. pseudotuberculosis T3SS expression is kept low under anaerobic, iron-rich conditions, such as those found in the intestinal lumen where the Yersinia T3SS is not required for growth. In contrast, the Yersinia T3SS is expressed under aerobic or anaerobic, iron-poor conditions, such as those encountered by Yersinia once they cross the epithelial barrier and encounter phagocytic cells. We further show that the [2Fe-2S] containing transcription factor, IscR, mediates this oxygen and iron regulation of the T3SS by controlling transcription of the T3SS master regulator LcrF. IscR binds directly to the lcrF promoter and, importantly, a mutation that prevents this binding leads to decreased disseminated infection of Y. pseudotuberculosis but does not perturb intestinal colonization. Similar to E. coli, Y. pseudotuberculosis uses the Fe-S cluster occupancy of IscR as a readout of oxygen and iron conditions that impact cellular Fe-S cluster homeostasis. We propose that Y. pseudotuberculosis has coopted this system to sense entry into deeper tissues and induce T3S where it is required for virulence. The IscR binding site in the lcrF promoter is completely conserved between Y. pseudotuberculosis and Y. pestis. Deletion of iscR in Y. pestis leads to drastic disruption of T3S, suggesting that IscR control of the T3SS evolved before Y. pestis split from Y. pseudotuberculosis.


Assuntos
Ferro/metabolismo , Oxigênio/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Infecções por Yersinia pseudotuberculosis/imunologia , Animais , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica/genética , Regiões Promotoras Genéticas/genética , Transcrição Gênica , Yersinia/metabolismo , Yersinia pseudotuberculosis/patogenicidade , Infecções por Yersinia pseudotuberculosis/metabolismo
9.
Microbiol Immunol ; 64(11): 768-777, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32902897

RESUMO

The Gram-negative bacterial pathogen Yersinia delivers six effector proteins into the host cells to block the host innate immune response. One of the effectors, YopT, is a potent cysteine protease that causes the disruption of the actin cytoskeleton to inhibit phagocytosis of the pathogen; however, its molecular mechanism and relevance to pathogenesis need further investigation. In this report, we show that RIG-I is a novel target of the YopT protein. Remarkably, YopT interacts with RIG-I and inhibits rat liver homogenate-mediated nuclear factor-κB and interferon regulatory factor-3 activation. Further studies revealed a YopT-dependent increase in the K48-polymerized ubiquitination of RIG-I. These findings suggest that YopT negatively regulates RIG-I-mediated cellular antibacterial response by targeting RIG-I.


Assuntos
Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Fator Regulador 3 de Interferon/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Yersinia/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Cisteína Endopeptidases/genética , Células HEK293 , Humanos , Camundongos , NF-kappa B/genética , Fagocitose , Células RAW 264.7 , Fator de Transcrição RelA , Yersinia/genética
10.
Infect Immun ; 87(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30602502

RESUMO

Pathogenic Yersinia species deliver Yop effector proteins through a type III secretion system into host cells. Among these effectors, YopE and YopT are Rho-modifying toxins, which function to modulate host cell physiology and evade immune responses. YopE is a GTPase-activating protein (GAP) while YopT is a protease, and they inhibit RhoA by different modes of action. Modifications to RhoA are sensed by pyrin, which, once activated, assembles a caspase-1 inflammasome, which generates cytokines such as interleukin-1ß (IL-1ß) and cell death by pyroptosis. In Yersinia-infected macrophages, YopE or YopT triggers inflammasome assembly only in the absence of another effector, YopM, which counteracts pyrin by keeping it inactive. The glucosyltransferase TcdB from Clostridium difficile, a well-studied RhoA-inactivating toxin, triggers activation of murine pyrin by dephosphorylation of Ser205 and Ser241. To determine if YopE or YopT triggers pyrin dephosphorylation, we infected lipopolysaccharide (LPS)-primed murine macrophages with ΔyopMYersinia pseudotuberculosis strains expressing wild-type (wt) or YopE mutant variants or YopT. By immunoblotting pyrin after infection, we observed that wt YopE triggered dephosphorylation of Ser205 and inflammasome activation. Pyrin dephosphorylation was reduced if a YopE variant had a defect in stability or RhoA specificity but not membrane localization. We also observed that wt YopT triggered pyrin dephosphorylation but more slowly than YopE, suggesting that YopE is dominant in this process. Our findings provide evidence that RhoA-modifying toxins trigger activation of pyrin by a conserved dephosphorylation mechanism. In addition, by characterization of YopE and YopT, we show that different features of effectors, such as RhoA specificity, affect the efficiency of pyrin dephosphorylation.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Bactérias/imunologia , Cisteína Endopeptidases/imunologia , Inflamassomos/metabolismo , Macrófagos/metabolismo , Pirina/metabolismo , Yersinia/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Macrófagos/imunologia , Fosforilação
11.
Int J Mol Sci ; 20(16)2019 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-31434224

RESUMO

Psychrotrophic foodborne pathogens, such as enteropathogenic Yersinia, which are able to survive and multiply at low temperatures, require cold shock proteins (Csps). The Csp superfamily consists of a diverse group of homologous proteins, which have been found throughout the eubacteria. They are related to cold shock tolerance and other cellular processes. Csps are mainly named following the convention of those in Escherichia coli. However, the nomenclature of certain Csps reflects neither their sequences nor functions, which can be confusing. Here, we performed phylogenetic analyses on Csp sequences in psychrotrophic enteropathogenic Yersinia and E. coli. We found that representative Csps in enteropathogenic Yersinia and E. coli can be clustered into six phylogenetic groups. When we extended the analysis to cover Enterobacteriales, the same major groups formed. Moreover, we investigated the evolutionary and structural relationships and the origin time of Csp superfamily members in eubacteria using nucleotide-level comparisons. Csps in eubacteria were classified into five clades and 12 subclades. The most recent common ancestor of Csp genes was estimated to have existed 3585 million years ago, indicating that Csps have been important since the beginning of evolution and have enabled bacterial growth in unfavorable conditions.


Assuntos
Proteínas de Bactérias/classificação , Proteínas de Bactérias/metabolismo , Proteínas e Peptídeos de Choque Frio/classificação , Proteínas e Peptídeos de Choque Frio/metabolismo , Escherichia coli/metabolismo , Eubacterium/metabolismo , Yersinia/metabolismo , Proteínas de Bactérias/genética , Proteínas e Peptídeos de Choque Frio/genética , Escherichia coli/genética , Eubacterium/genética , Filogenia , Yersinia/genética
12.
J Biol Chem ; 292(19): 8092-8100, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28280241

RESUMO

Pathogenic Yersinia bacteria cause a range of human diseases. To modulate and evade host immune systems, these yersiniae inject effector proteins into host macrophages. One such protein, the serine/threonine kinase YopO (YpkA in Yersinia pestis), uses monomeric actin as bait to recruit and phosphorylate host actin polymerization-regulating proteins, including the actin-severing protein gelsolin, to disrupt actin filaments and thus impair phagocytosis. However, the YopO phosphorylation sites on gelsolin and the consequences of YopO-mediated phosphorylation on actin remodeling have yet to be established. Here we determined the effects of YopO-mediated phosphorylation on gelsolin and identified its phosphorylation sites by mass spectrometry. YopO phosphorylated gelsolin in the linker region between gelsolin homology domains G3 and G4, which, in the absence of calcium, are compacted but adopt an open conformation in the presence of calcium, enabling actin binding and severing. Using phosphomimetic and phosphodeletion gelsolin mutants, we found that YopO-mediated phosphorylation partially mimics calcium-dependent activation of gelsolin, potentially contributing to a reduction in filamentous actin and altered actin dynamics in phagocytic cells. In summary, this work represents the first report of the functional outcome of serine/threonine phosphorylation in gelsolin regulation and provides critical insight into how YopO disrupts normal gelsolin function to alter host actin dynamics and thus cripple phagocytosis.


Assuntos
Actinas/química , Proteínas de Bactérias/metabolismo , Cálcio/química , Gelsolina/química , Proteínas Serina-Treonina Quinases/metabolismo , Yersinia/metabolismo , Citoesqueleto de Actina/metabolismo , Sítios de Ligação , Humanos , Macrófagos/microbiologia , Espectrometria de Massas , Simulação de Dinâmica Molecular , Mutação , Fagocitose , Fosforilação , Domínios Proteicos , Pirenos/química , Serina/química , Treonina/química
13.
Curr Top Microbiol Immunol ; 399: 201-220, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27744508

RESUMO

Pathogenic bacteria of the genus Yersinia include Y. pestis-the agent of plaque-and two enteropathogens, Y. enterocolitica, and Y. pseudotuberculosis. These pathogens have developed an array of virulence factors aimed at manipulating Rho GTP-binding proteins and the actin cytoskeleton in host cells to cross the intestinal barrier and suppress the immune system. Yersinia virulence factors include outer membrane proteins triggering cell invasion by binding to integrins, effector proteins injected into host cells to manipulate Rho protein functions and a Rho protein-activating exotoxin. Here, we present an overview of how Yersinia and host factors are integrated in a regulatory network that orchestrates the subversion of host defense.


Assuntos
Actinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Yersiniose/enzimologia , Yersinia/metabolismo , Quinases Associadas a rho/metabolismo , Actinas/genética , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Yersinia/genética , Yersiniose/metabolismo , Yersiniose/microbiologia , Quinases Associadas a rho/genética
14.
Mol Microbiol ; 102(4): 593-610, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27507539

RESUMO

Three pathogenic species of the genus Yersinia assemble adhesive fimbriae via the FGL-chaperone/usher pathway. Closely related Y. pestis and Y. pseudotuberculosis elaborate the pH6 antigen (Psa), which mediates bacterial attachment to alveolar cells of the lung. Y. enterocolitica, instead, assembles the homologous fimbriae Myf of unknown function. Here, we discovered that Myf, like Psa, specifically recognizes ß1-3- or ß1-4-linked galactose in glycosphingolipids, but completely lacks affinity for phosphatidylcholine, the main receptor for Psa in alveolar cells. The crystal structure of a subunit of Psa (PsaA) complexed with choline together with mutagenesis experiments revealed that PsaA has four phosphatidylcholine binding pockets that enable super-high-avidity binding of Psa-fibres to cell membranes. The pockets are arranged as six tyrosine residues, which are all missing in the MyfA subunit of Myf. Conversely, the crystal structure of the MyfA-galactose complex revealed that the galactose-binding site is more extended in MyfA, enabling tighter binding to lactosyl moieties. Our results suggest that during evolution, Psa has acquired a tyrosine-rich surface that enables it to bind to phosphatidylcholine and mediate adhesion of Y. pestis/pseudotuberculosis to alveolar cells, whereas Myf has specialized as a carbohydrate-binding adhesin, facilitating the attachment of Y. enterocolitica to intestinal cells.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Fímbrias Bacterianas/metabolismo , Yersinia/metabolismo , Adesinas Bacterianas/metabolismo , Sequência de Aminoácidos , Antígenos de Bactérias/genética , Antígenos de Bactérias/ultraestrutura , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/ultraestrutura , Sítios de Ligação , Proteínas de Fímbrias/metabolismo , Chaperonas Moleculares/metabolismo , Tropismo/genética , Virulência/genética , Yersinia enterocolitica/metabolismo , Yersinia pestis/metabolismo , Yersinia pseudotuberculosis/metabolismo
15.
Proc Natl Acad Sci U S A ; 111(18): 6768-73, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753568

RESUMO

The genus Yersinia has been used as a model system to study pathogen evolution. Using whole-genome sequencing of all Yersinia species, we delineate the gene complement of the whole genus and define patterns of virulence evolution. Multiple distinct ecological specializations appear to have split pathogenic strains from environmental, nonpathogenic lineages. This split demonstrates that contrary to hypotheses that all pathogenic Yersinia species share a recent common pathogenic ancestor, they have evolved independently but followed parallel evolutionary paths in acquiring the same virulence determinants as well as becoming progressively more limited metabolically. Shared virulence determinants are limited to the virulence plasmid pYV and the attachment invasion locus ail. These acquisitions, together with genomic variations in metabolic pathways, have resulted in the parallel emergence of related pathogens displaying an increasingly specialized lifestyle with a spectrum of virulence potential, an emerging theme in the evolution of other important human pathogens.


Assuntos
Evolução Molecular , Virulência/genética , Yersinia/genética , Yersinia/patogenicidade , Genoma Bacteriano , Humanos , Redes e Vias Metabólicas/genética , Filogenia , Especificidade da Espécie , Yersinia/metabolismo , Yersinia enterocolitica/genética , Yersinia enterocolitica/metabolismo , Yersinia enterocolitica/patogenicidade
16.
J Biol Chem ; 290(31): 18967-74, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26055720

RESUMO

Numerous pathogenic microorganisms secrete small molecule chelators called siderophores defined by their ability to bind extracellular ferric iron, making it bioavailable to microbes. Recently, a siderophore produced by uropathogenic Escherichia coli, yersiniabactin, was found to also bind copper ions during human infections. The ability of yersiniabactin to protect E. coli from copper toxicity and redox-based phagocyte defenses distinguishes it from other E. coli siderophores. Here we compare yersiniabactin to other extracellular copper-binding molecules and review how copper-binding siderophores may confer virulence-associated gains of function during infection pathogenesis.


Assuntos
Cobre/fisiologia , Interações Hospedeiro-Patógeno , Sideróforos/fisiologia , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Escherichia coli/imunologia , Escherichia coli/metabolismo , Humanos , Imunidade Inata , Yersinia/imunologia , Yersinia/metabolismo
17.
Infect Immun ; 84(8): 2243-2254, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27217422

RESUMO

Leucine-rich repeat (LRR) proteins are widely distributed in bacteria, playing important roles in various protein-protein interaction processes. In Yersinia, the well-characterized type III secreted effector YopM also belongs to the LRR protein family and is encoded by virulence plasmids. However, little has been known about other LRR members encoded by Yersinia genomes or their evolution. In this study, the Yersinia LRR proteins were comprehensively screened, categorized, and compared. The LRR proteins encoded by chromosomes (LRR1 proteins) appeared to be more similar to each other and different from those encoded by plasmids (LRR2 proteins) with regard to repeat-unit length, amino acid composition profile, and gene expression regulation circuits. LRR1 proteins were also different from LRR2 proteins in that the LRR1 proteins contained an E3 ligase domain (NEL domain) in the C-terminal region or an NEL domain-encoding nucleotide relic in flanking genomic sequences. The LRR1 protein-encoding genes (LRR1 genes) varied dramatically and were categorized into 4 subgroups (a to d), with the LRR1a to -c genes evolving from the same ancestor and LRR1d genes evolving from another ancestor. The consensus and ancestor repeat-unit sequences were inferred for different LRR1 protein subgroups by use of a maximum parsimony modeling strategy. Structural modeling disclosed very similar repeat-unit structures between LRR1 and LRR2 proteins despite the different unit lengths and amino acid compositions. Structural constraints may serve as the driving force to explain the observed mutations in the LRR regions. This study suggests that there may be functional variation and lays the foundation for future experiments investigating the functions of the chromosomally encoded LRR proteins of Yersinia.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Evolução Molecular , Proteínas/genética , Proteínas/metabolismo , Yersinia/genética , Yersinia/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/química , Sequência Consenso , Regulação Bacteriana da Expressão Gênica , Variação Genética , Genoma Bacteriano , Proteínas de Repetições Ricas em Leucina , Modelos Moleculares , Fases de Leitura Aberta , Filogenia , Matrizes de Pontuação de Posição Específica , Conformação Proteica , Transporte Proteico , Proteínas/química , Análise de Sequência de DNA
18.
Biochim Biophys Acta ; 1851(6): 911-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25241942

RESUMO

Phosphoinositides control key cellular processes including vesicular trafficking and actin polymerization. Intracellular bacterial pathogens manipulate phosphoinositide metabolism in order to promote their uptake by target cells and to direct in some cases the biogenesis of their replication compartments. In this chapter, we review the molecular strategies that major pathogens including Listeria, Mycobacterium, Shigella, Salmonella, Legionella and Yersinia use to hijack phosphoinositides during infection. This article is part of a Special Issue entitled Phosphoinositides.


Assuntos
Infecções Bacterianas/metabolismo , Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Fosfatidilinositóis/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Expressão Gênica , Humanos , Legionella/imunologia , Legionella/metabolismo , Listeria/imunologia , Listeria/metabolismo , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/metabolismo , Fosfatidilinositóis/imunologia , Ligação Proteica , Salmonella/imunologia , Salmonella/metabolismo , Shigella/imunologia , Shigella/metabolismo , Vesículas Transportadoras/metabolismo , Yersinia/imunologia , Yersinia/metabolismo
19.
Mol Microbiol ; 95(1): 80-100, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25353290

RESUMO

Intimin and Invasin are prototypical inverse (Type Ve) autotransporters and important virulence factors of enteropathogenic Escherichia coli and Yersinia spp. respectively. In addition to a C-terminal extracellular domain and a ß-barrel transmembrane domain, both proteins also contain a short N-terminal periplasmic domain that, in Intimin, includes a lysin motif (LysM), which is thought to mediate binding to peptidoglycan. We show that the periplasmic domain of Intimin does bind to peptidoglycan both in vitro and in vivo, but only under acidic conditions. We were able to determine a dissociation constant of 0.8 µM for this interaction, whereas the Invasin periplasmic domain, which lacks a LysM, bound only weakly in vitro and failed to bind peptidoglycan in vivo. We present the solution structure of the Intimin LysM, which has an additional α-helix conserved within inverse autotransporter LysMs but lacking in others. In contrast to previous reports, we demonstrate that the periplasmic domain of Intimin mediates dimerisation. We further show that dimerisation and peptidoglycan binding are general features of LysM-containing inverse autotransporters. Peptidoglycan binding by the periplasmic domain in the infection process may aid in resisting mechanical and chemical stress during transit through the gastrointestinal tract.


Assuntos
Adesinas Bacterianas/química , Adesinas Bacterianas/metabolismo , Escherichia coli Enteropatogênica/metabolismo , Peptidoglicano/metabolismo , Yersinia/metabolismo , Adesinas Bacterianas/genética , Sítios de Ligação , Biologia Computacional/métodos , Dimerização , Escherichia coli Enteropatogênica/química , Escherichia coli Enteropatogênica/genética , Concentração de Íons de Hidrogênio , Modelos Moleculares , Multimerização Proteica , Estrutura Secundária de Proteína , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Yersinia/química , Yersinia/genética
20.
J Bacteriol ; 198(4): 604-14, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26644429

RESUMO

Many Gram-negative pathogens express a type III secretion (T3SS) system to enable growth and survival within a host. The three human-pathogenic Yersinia species, Y. pestis, Y. pseudotuberculosis, and Y. enterocolitica, encode the Ysc T3SS, whose expression is controlled by an AraC-like master regulator called LcrF. In this review, we discuss LcrF structure and function as well as the environmental cues and pathways known to regulate LcrF expression. Similarities and differences in binding motifs and modes of action between LcrF and the Pseudomonas aeruginosa homolog ExsA are summarized. In addition, we present a new bioinformatics analysis that identifies putative LcrF binding sites within Yersinia target gene promoters.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Transativadores/metabolismo , Sistemas de Secreção Tipo III/genética , Yersinia/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Estrutura Terciária de Proteína , Transativadores/química , Transativadores/genética , Sistemas de Secreção Tipo III/metabolismo , Yersinia/química , Yersinia/genética , Yersinia/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA