Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Immunol ; 205(7): 1878-1885, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32839236

RESUMO

Placental immune responses are highly regulated to strike a balance between protection and tolerance. For relatively mild infections, protection encompasses both the mother and fetus; however, during worsening conditions, protection becomes exclusively reserved for the mother. Previously, we and others have shown that the host factor perforin-2 plays a central role in protecting mice and cells against infection. In this study, we analyzed perforin-2 activity in the mouse placenta to determine whether perforin-2 plays a similarly protective role. We show that perforin-2 is critical for inhibiting Listeria monocytogenes colonization of the placenta and fetus and that this protection is due to both maternal and fetal-encoded perforin-2. Perforin-2 mRNA is readily detectable in individual immune cells of the decidua, and these levels are further enhanced specifically in decidual macrophages during high-dose infections that result in fetal expulsion. Unexpectedly, inductive perforin-2 expression in decidual macrophages did not occur during milder infections in which fetal viability remained intact. This pattern of expression significantly differed from that observed in splenic macrophages in which inductive perforin-2 expression was observed in both high and mild infection conditions. In the placenta, inductive perforin-2 expression in decidual macrophages was coincident with their polarization from a CD206+ MHC class IIlo to CD206- MHC class IIhi phenotype that normally occurs in the placenta during high-burden infections. Our results suggest that perforin-2 is part of a host response that is protective either for both the mother and fetus in milder infections or exclusively for the mother during high-dose infections.


Assuntos
Feto/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Placenta/imunologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Complicações Infecciosas na Gravidez/imunologia , Animais , Patógenos Transmitidos pelo Sangue , Células Cultivadas , Feminino , Humanos , Imunidade Materno-Adquirida , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/microbiologia , Proteínas Citotóxicas Formadoras de Poros/genética , Gravidez , Análise de Célula Única
2.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29311243

RESUMO

The host employs both cell-autonomous and system-level responses to limit pathogen replication in the initial stages of infection. Previously, we reported that the eukaryotic initiation factor 2α (eIF2α) kinases heme-regulated inhibitor (HRI) and protein kinase R (PKR) control distinct cellular and immune-related activities in response to diverse bacterial pathogens. Specifically for Listeria monocytogenes, there was reduced translocation of the pathogen to the cytosolic compartment in HRI-deficient cells and consequently reduced loading of pathogen-derived antigens on major histocompatibility complex class I (MHC-I) complexes. Here we show that Hri-/- mice, as well as wild-type mice treated with an HRI inhibitor, are more susceptible to listeriosis. In the first few hours of L. monocytogenes infection, there was much greater pathogen proliferation in the liver of Hri-/- mice than in the liver of Hri+/+ mice. Further, there was a rapid increase of serum interleukin-6 (IL-6) levels in Hri+/+ mice in the first few hours of infection whereas the increase in IL-6 levels in Hri-/- mice was notably delayed. Consistent with these in vivo findings, the rate of listeriolysin O (LLO)-dependent pathogen efflux from infected Hri-/- macrophages and fibroblasts was significantly higher than the rate seen with infected Hri+/+ cells. Treatment of cells with an eIF2α kinase activator enhanced both the HRI-dependent and PKR-dependent infection phenotypes, further indicating the pharmacologically malleability of this signaling pathway. Collectively, these results suggest that HRI mediates the cellular confinement and killing of virulent L. monocytogenes in addition to promoting a system-level cytokine response and that both are required to limit pathogen replication during the first few hours of infection.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/enzimologia , Proteínas Serina-Treonina Quinases/imunologia , Animais , Feminino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Listeria monocytogenes/genética , Listeriose/genética , Listeriose/imunologia , Listeriose/microbiologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteínas Serina-Treonina Quinases/genética
3.
Infect Immun ; 84(4): 1083-1091, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26831467

RESUMO

The host-encoded Perforin-2 (encoded by the macrophage-expressed gene 1, Mpeg1), which possesses a pore-forming MACPF domain, reduces the viability of bacterial pathogens that reside within membrane-bound compartments. Here, it is shown that Perforin-2 also restricts the proliferation of the intracytosolic pathogen Listeria monocytogenes Within a few hours of systemic infection, the massive proliferation of L. monocytogenes in Perforin-2(-/-)mice leads to a rapid appearance of acute disease symptoms. We go on to show in cultured Perforin-2(-/-)cells that the vacuole-to-cytosol transitioning of L. monocytogenesis greatly accelerated. Unexpectedly, we found that in Perforin-2(-/-)macrophages,Listeria-containing vacuoles quickly (≤ 15 min) acidify, and that this was coincident with greater virulence gene expression, likely accounting for the more rapid translocation of L. monocytogenes to its replicative niche in the cytosol. This hypothesis was supported by our finding that aL. monocytogenes strain expressing virulence factors at a constitutively high level replicated equally well in Perforin-2(+/+)and Perforin-2(-/-)macrophages. Our findings suggest that the protective role of Perforin-2 against listeriosis is based on it limiting the intracellular replication of the pathogen. This cellular activity of Perforin-2 may derive from it regulating the acidification of Listeria-containing vacuoles, thereby depriving the pathogen of favorable intracellular conditions that promote its virulence gene activity.


Assuntos
Citosol/fisiologia , Listeria monocytogenes/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Vacúolos/fisiologia , Animais , Células Cultivadas , Citosol/microbiologia , Regulação da Expressão Gênica/fisiologia , Listeria monocytogenes/ultraestrutura , Listeriose/metabolismo , Listeriose/microbiologia , Proteínas de Membrana/genética , Camundongos , Proteínas Citotóxicas Formadoras de Poros/genética , Estrutura Terciária de Proteína
4.
Infect Immun ; 83(11): 4404-15, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26351279

RESUMO

Induction of adaptive immunity leads to the establishment of immunological memory; however, how innate immunity regulates memory T cell function remains obscure. Here we show a previously undefined mechanism in which innate and adaptive immunity are linked by TIR domain-containing adapter-inducing beta interferon (TRIF) during establishment and reactivation of memory T cells against Gram-negative enteropathogens. Absence of TRIF in macrophages (Mϕs) but not dendritic cells led to a predominant generation of CD4(+) central memory T cells that express IL-17 during enteric bacterial infection in mice. TRIF-dependent type I interferon (IFN) signaling in T cells was essential to Th1 lineage differentiation and reactivation of memory T cells. TRIF activated memory T cells to facilitate local neutrophil influx and enhance bacterial elimination. These results highlight the importance of TRIF as a mediator of the innate and adaptive immune interactions in achieving the protective properties of memory immunity against Gram-negative bacteria and suggest TRIF as a potential therapeutic target.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Memória Imunológica , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Células Dendríticas/imunologia , Humanos , Interferon gama/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Yersiniose/genética , Yersiniose/microbiologia , Yersinia enterocolitica/genética
5.
Infect Immun ; 82(2): 762-72, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478090

RESUMO

Neonatal animals are generally very susceptible to infection with bacterial pathogens. However, we recently reported that neonatal mice are highly resistant to orogastric infection with Yersinia enterocolitica. Here, we show that proinflammatory responses greatly exceeding those in adults arise very rapidly in the mesenteric lymph nodes (MLN) of neonates. High-level induction of proinflammatory gene expression occurred in the neonatal MLN as early as 18 h postinfection. Marked innate phagocyte recruitment was subsequently detected at 24 h postinfection. Enzyme-linked immunosorbent spot assay (ELISPOT) analyses indicated that enhanced inflammation in neonatal MLN is contributed to, in part, by an increased frequency of proinflammatory cytokine-secreting cells. Moreover, both CD11b(+) and CD11b(-) cell populations appeared to play a role in proinflammatory gene expression. The level of inflammation in neonatal MLN was also dependent on key bacterial components. Y. enterocolitica lacking the virulence plasmid failed to induce innate phagocyte recruitment. In contrast, tumor necrosis factor alpha (TNF-α) protein expression and neutrophil recruitment were strikingly higher in neonatal MLN after infection with a yopP-deficient strain than with wild-type Y. enterocolitica, whereas only modest increases occurred in adults. This hyperinflammatory response was associated with greater colonization of the spleen and higher mortality in neonates, while there was no difference in mortality among adults. This model highlights the dynamic levels of inflammation in the intestinal lymphoid tissues and reveals the protective (wild-type strain) versus harmful (yopP-deficient strain) consequences of inflammation in neonates. Moreover, these results reveal that the neonatal intestinal lymphoid tissues have great potential to rapidly mobilize innate components in response to infection with bacterial enteropathogens.


Assuntos
Linfonodos/imunologia , Linfonodos/microbiologia , Linfadenite Mesentérica/imunologia , Linfadenite Mesentérica/microbiologia , Yersiniose/imunologia , Yersiniose/patologia , Yersinia enterocolitica/imunologia , Animais , Animais Recém-Nascidos , Perfilação da Expressão Gênica , Inflamação , Linfonodos/patologia , Macrófagos/imunologia , Linfadenite Mesentérica/patologia , Camundongos , Baço/microbiologia , Análise de Sobrevida , Yersiniose/microbiologia
6.
J Biol Chem ; 287(34): 28738-44, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22761422

RESUMO

In eukaryotic cells, there are two well characterized pathways that regulate translation initiation in response to stress, and each have been shown to be targeted by various viruses. We recently showed in a yeast-based model that the bacterial virulence factor YopJ disrupts one of these pathways, which is centered on the α-subunit of the translation factor eIF2. Here, we show in mammalian cells that induction of the eIF2 signaling pathway occurs following infection with bacterial pathogens and that, consistent with our yeast-based findings, YopJ reduces eIF2 signaling in response to endoplasmic reticulum stress, heavy metal toxicity, dsRNA, and bacterial infection. We demonstrate that the well documented activities of YopJ, inhibition of NF-κB activation and proinflammatory cytokine expression, are both dependent on an intact eIF2 signaling pathway. Unexpectedly, we found that cells with defective eIF2 signaling were more susceptible to bacterial invasion. This was true for pathogenic Yersinia, a facultative intracellular pathogen, as well as for the intracellular pathogens Listeria monocytogenes and Chlamydia trachomatis. Collectively, our data indicate that the highly conserved eIF2 signaling pathway, which is vitally important for antiviral responses, plays a variety of heretofore unrecognized roles in antibacterial responses.


Assuntos
Infecções por Chlamydia/metabolismo , Chlamydia trachomatis/metabolismo , Citocinas/biossíntese , Fator de Iniciação 2 em Eucariotos/metabolismo , Mediadores da Inflamação/metabolismo , Listeria monocytogenes/metabolismo , Listeriose/metabolismo , Transdução de Sinais , Yersiniose/metabolismo , Yersinia/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Linhagem Celular , Infecções por Chlamydia/genética , Infecções por Chlamydia/imunologia , Chlamydia trachomatis/genética , Chlamydia trachomatis/imunologia , Citocinas/genética , Citocinas/imunologia , Fator de Iniciação 2 em Eucariotos/genética , Fator de Iniciação 2 em Eucariotos/imunologia , Mediadores da Inflamação/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/imunologia , Listeriose/genética , Listeriose/imunologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , NF-kappa B/metabolismo , Yersinia/genética , Yersinia/imunologia , Yersiniose/genética , Yersiniose/imunologia
7.
Microb Pathog ; 52(1): 41-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22023991

RESUMO

The plague bacterium Yersinia pestis has a number of well-described strategies to protect itself from both host cells and soluble factors. In an effort to identify additional anti-host factors, we employed a transposon site hybridization (TraSH)-based approach to screen 10(5)Y. pestis mutants in an in vitro infection system. In addition to loci encoding various components of the well-characterized type III secretion system (T3SS), our screen unambiguously identified ompA as a pro-survival gene. We go on to show that an engineered Y. pestis ΔompA strain, as well as a ΔompA strain of the closely related pathogen Yersinia pseudotuberculosis, have fully functioning T3SSs but are specifically defective in surviving within macrophages. Additionally, the Y. pestis ΔompA strain was out competed by the wild-type strain in a mouse co-infection assay. Unlike in other bacterial pathogens in which OmpA can promote adherence, invasion, or serum resistance, the OmpA of Y. pestis is restricted to enhancing intracellular survival. Our data show that OmpA of the pathogenic Yersinia is a virulence factor on par with the T3SS.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Viabilidade Microbiana , Peste/microbiologia , Yersinia pestis/metabolismo , Yersinia pestis/patogenicidade , Animais , Proteínas da Membrana Bacteriana Externa/genética , Feminino , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Virulência , Yersinia pestis/genética , Yersinia pestis/crescimento & desenvolvimento
8.
J Biol Chem ; 284(37): 24744-53, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19553678

RESUMO

The Yersinia protein kinase A (YpkA) and outer protein J (YopJ) are co-expressed from a single transcript and are injected directly into eukaryotic cells by the plague bacterium Yersinia pestis. When overexpressed in vertebrate or yeast cells, YpkA disrupts the actin-based cytoskeletal system by an unknown mechanism, whereas YopJ obstructs inductive chemokine expression by inhibiting MAPK and NF-kappaB signaling. Previously, we showed that the fission yeast Schizosaccharomyces pombe was sensitive to the kinase activity of YpkA. Here, we screened yeast for cellular processes important for YpkA activity and found that the eIF2alpha kinases mollify the toxicity imparted by the kinase activity of YpkA. Specifically, strains lacking the eIF2alpha kinase Hri2 were particularly sensitive to YpkA. Unexpectedly, the activity of YopJ, which conferred a phenotype consistent with its inhibitory effect on MAPK signaling, was also found to be dependent on Hri2. When expressed in S. pombe, YopJ sensitized cells to osmotic and oxidative stresses through a Hri2-dependent mechanism. However, when co-expressed with YpkA, YopJ protected cells from YpkA-mediated toxicity, and this protection was entirely dependent on Hri2. In contrast, YopJ did not confer protection against the toxic effects of the Yersinia virulence factor YopE. These findings are the first to functionally link YpkA and YopJ and suggest that eIF2alpha kinases, which are critically important in antiviral defenses and protection against environmental stresses, also play a role in bacterial virulence.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Linhagem Celular , Sistema de Sinalização das MAP Quinases , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Modelos Biológicos , Plasmídeos/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Schizosaccharomyces/enzimologia , Fatores de Tempo , Yersinia pestis/metabolismo , Yersinia pseudotuberculosis/metabolismo
9.
Infect Immun ; 78(8): 3595-608, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20515925

RESUMO

Mucosal immunity to gastrointestinal pathogens in early life has been studied only slightly. Recently, we developed an infection model in murine neonates using the gastroenteric pathogen Yersinia enterocolitica. Here, we report that oral infection of neonatal mice with low doses of virulent Y. enterocolitica leads to vigorous intestinal and systemic adaptive immunity. Y. enterocolitica infection promoted the development of anti-LcrV memory serum IgG1 and IgG2a responses of comparable affinity and magnitude to adult responses. Strikingly, neonatal mesenteric lymph node CD4(+) T cells produced Yersinia-specific gamma interferon (IFN-gamma) and interleukin-17A (IL-17A), exceeding adult levels. The robust T- and B-cell responses elicited in neonates exposed to Y. enterocolitica were associated with long-term protection against mucosal challenge with this pathogen. Using genetically deficient mice, we found that IFN-gamma and CD4(+) cells, but not B cells, are critical for protection of neonates during primary Y. enterocolitica infection. In contrast, adults infected with low bacterial doses did not require either cell population for protection. CD4-deficient neonatal mice adoptively transferred with CD4(+) cells from wild-type, IFN-gamma-deficient, or IL-17AF-deficient mice were equally protected from infection. These data demonstrate that inflammatory CD4(+) T cells are required for protection of neonatal mice and that this protection may not require CD4-derived IFN-gamma, IL-17A, or IL-17F. Overall, these studies support the idea that Y. enterocolitica promotes the development of highly inflammatory mucosal responses in neonates and that intestinal T-cell function may be a key immune component in protection from gastrointestinal pathogens in early life.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Linfócitos B/imunologia , Feminino , Imunidade Inata , Imunoglobulina G/sangue , Interferon gama/metabolismo , Interleucina-17/metabolismo , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/imunologia
10.
J Vis Exp ; (144)2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30855571

RESUMO

There are a variety of strategies bacterial pathogens employ to survive and proliferate once inside the eukaryotic cell. The so-called 'cytosolic' pathogens (Listeria monocytogenes, Shigella flexneri, Burkholderia pseudomallei, Francisella tularensis, and Rickettsia spp.) gain access to the infected cell cytosol by physically and enzymatically degrading the primary vacuolar membrane. Once in the cytosol, these pathogens both proliferate as well as generate sufficient mechanical forces to penetrate the plasma membrane of the host cell in order to infect new cells. Here, we show how this terminal step of the cellular infection cycle of L. monocytogenes (Lm) can be quantified by both colony-forming unit assays and flow cytometry and give examples of how both pathogen- and host-encoded factors impact this process. We also show a close correspondence of Lm infection dynamics of cultured cells infected in vitro and those of hepatic cells derived from mice infected in vivo. These function-based assays are relatively simple and can be readily scaled up for discovery-based high-throughput screens for modulators of eukaryotic cell function.


Assuntos
Fenômenos Fisiológicos Bacterianos , Interações Hospedeiro-Patógeno , Animais , Membrana Celular/metabolismo , Membrana Celular/microbiologia , Citosol/metabolismo , Citosol/microbiologia , Humanos , Camundongos
11.
Artigo em Inglês | MEDLINE | ID: mdl-30854334

RESUMO

Yersinia pestis, the causative agent of plague, possesses a number of virulence mechanisms that allows it to survive and proliferate during its interaction with the host. To discover additional infection-specific Y. pestis factors, a transposon site hybridization (TraSH)-based genome-wide screen was employed to identify genomic regions required for its survival during cellular infection. In addition to several well-characterized infection-specific genes, this screen identified three chromosomal genes (y3397, y3399, and y3400), located in an apparent operon, that promoted successful infection. Each of these genes is predicted to encode a leucine-rich repeat family protein with or without an associated ubiquitin E3 ligase domain. These genes were designated Yersinia leucine-rich repeat gene A (ylrA), B (ylrB), and C (ylrC). Engineered strains with deletions of y3397 (ylrC), y3399 (ylrB), or y3400 (ylrA), exhibited infection defects both in cultured cells and in the mouse. C-terminal FLAG-tagged YlrA, YlrB, and YlrC were secreted by Y. pestis in the absence but not the presence of extracellular calcium and deletions of the DNA sequences encoding the predicted N-terminal type III secretion signals of YlrA, YlrB, and YlrC prevented their secretion, indicating that these proteins are substrates of the type III secretion system (T3SS). Further strengthening the connection with the T3SS, YlrB was readily translocated into HeLa cells and expression of the YlrA and YlrC proteins in yeast inhibited yeast growth, indicating that these proteins may function as anti-host T3S effector proteins.


Assuntos
Interações Hospedeiro-Patógeno , Peste/fisiopatologia , Sistemas de Secreção Tipo III/metabolismo , Fatores de Virulência/metabolismo , Yersinia pestis/patogenicidade , Animais , Transporte Biológico , Cromossomos Bacterianos , Modelos Animais de Doenças , Deleção de Genes , Genes Bacterianos , Testes Genéticos , Células HeLa , Humanos , Camundongos , Modelos Teóricos , Células RAW 264.7 , Virulência , Fatores de Virulência/genética , Yersinia pestis/genética
12.
J Bacteriol ; 190(10): 3774-8, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18359811

RESUMO

Yersinia spp. use a type 3 secretion system (T3SS) to directly inject six proteins into macrophages, and any impairment of this process results in a profound reduction in virulence. We previously showed that the exoribonuclease polynucleotide phosphorylase (PNPase) was required for optimal T3SS functioning in Yersinia pseudotuberculosis and Yersinia pestis. Here we report that Y. pseudotuberculosis cells with reduced RNase E activity are likewise impaired in T3SS functioning and that phenotypically they resemble Delta pnp cells. RNase E does not affect expression levels of the T3SS substrates but instead, like PNPase, regulates a terminal event in the secretion pathway. This similarity, together with the fact that RNase E and PNPase can be readily copurified from Y. pseudotuberculosis cell extracts, suggests that these two RNases regulate T3SS activity through a common mechanism. This is the first report that RNase E activity impacts the T3SS as well as playing a more general role in infectivity.


Assuntos
Endorribonucleases/fisiologia , Transporte Proteico , Fatores de Virulência/metabolismo , Yersinia/metabolismo , Endorribonucleases/genética , Regulação Bacteriana da Expressão Gênica , Virulência/genética , Fatores de Virulência/genética , Yersinia/enzimologia , Yersinia/fisiologia , Yersiniose/microbiologia
13.
J Mol Biol ; 373(1): 27-37, 2007 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-17825321

RESUMO

Pathogenic Yersinia spp. possess a protein secretion system, designated as type 3, that plays a clear role in promoting their survival vis-à-vis the macrophage. Inductive expression of the Yersinia type 3 secretion system (T3SS), triggered either by host cell contact, or, in the absence of host cells, by a reduction in extracellular calcium ion levels, is accompanied by a withdrawal from the bacterial division cycle. Here, we analyzed Ca(2+)-dependent induction of the T3SS at the single-cell level to understand how Yersinia coordinates pro-survival and growth-related activities. We utilized a novel high-throughput quantitative microscopy approach as well as flow cytometry to determine how Ca(2+) levels, T3SS expression, and cellular division are interrelated. Our analysis showed that there is a high degree of homogeneity in terms of T3SS expression levels among a population of Y. pseudotuberculosis cells following the removal of Ca(2+), and that T3SS expression appears to be independent of the cellular division cycle. Unexpectedly, our analysis showed that Ca(2+) levels are inversely related to the initiation of inductive T3SS expression, and not to the intensity of activation once initiated, thus providing a basis for the seemingly graded response of T3SS activation observed in bulk-level analyses. The properties of the system described here display both similarities to and differences from that of the lac operon first described 50 years ago by Novick and Weiner.


Assuntos
Proteínas de Bactérias/metabolismo , Divisão Celular/fisiologia , Yersinia/fisiologia , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Cálcio/metabolismo , Citometria de Fluxo/métodos , Regulação Bacteriana da Expressão Gênica , Microscopia/métodos , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Yersinia/citologia
14.
FEMS Microbiol Lett ; 270(2): 255-64, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17391372

RESUMO

Previously, it was shown that optimal functioning of the Yersinia type III secretion system (T3SS) in cell culture infection assays requires the exoribonuclease polynucleotide phosphorylase (PNPase) and that normal T3SS activity could be restored in the Deltapnp strains by expressing just the approximately 70-aa S1 RNA-binding domain of PNPase. Here, it is shown that the Yersinia Deltapnp strain is less virulent in the mouse compared with the isogenic wild-type strain. To begin to understand what could be limiting T3SS activity in the absence of PNPase, T3SS-encoding transcripts and proteins in the YersiniaDeltapnp strains were analyzed. Surprisingly, it was found that the Deltapnp Yersinia strains possessed enhanced levels of T3SS-encoding transcripts and proteins compared with the wild-type strains. We then found that an S1 variant containing a disruption in its RNA-binding subdomain was inactive in terms of restoring normal T3SS activity. However, T3SS expression levels did not differ between Deltapnp strains expressing active and inactive S1 proteins, further showing that T3SS activity and expression levels, at least as related to PNPase and its S1 domain, are not linked. The results suggest that PNPase affects the expression and activity of the T3SS by distinct mechanisms and that the S1-dependent effect on T3SS activity involves an RNA intermediate.


Assuntos
Proteínas de Bactérias/metabolismo , Polirribonucleotídeo Nucleotidiltransferase/metabolismo , Fatores de Virulência/metabolismo , Yersinia/enzimologia , Animais , Proteínas de Bactérias/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Polirribonucleotídeo Nucleotidiltransferase/genética , Transporte Proteico , Virulência/genética , Fatores de Virulência/genética , Yersinia/genética , Yersinia/patogenicidade , Yersiniose/microbiologia
15.
Adv Exp Med Biol ; 603: 217-24, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17966418

RESUMO

Low temperatures as well as encounters with host phagocytes are two stresses that have been relatively well studied in many species of bacteria. The exoribonuclease polynucleotide phosphorylase (PNPase) has previously been shown to be required by several species of bacteria, including Yersinia, for low-temperature growth. We have shown that PNPase also enhances the ability of Yersinia to withstand the killing activities of murine macrophages. We have gone on to show that PNPase is required for the optimal functioning of Yersinia's type three secretion system (T3SS), an organelle that injects effector proteins directly into host cells. Surprisingly, the PNPase-mediated effect on T3SS activity is independent of PNPase's ribonuclease activity and instead requires only its S1 RNA-binding domain. In stark contrast, the catalytic activity of PNPase is strictly required for enhanced growth at low temperature. Preliminary experiments suggest that the RNA-binding interface of the S1 domain is critical for its T3SS-enhancing activity. Our findings indicate that PNPase plays versatile roles in promoting Yersinia's survival in response to stressful conditions.


Assuntos
Polirribonucleotídeo Nucleotidiltransferase/fisiologia , Yersinia/fisiologia , Animais , Genes Bacterianos , Humanos , Macrófagos/imunologia , Camundongos , Mutação , Polirribonucleotídeo Nucleotidiltransferase/genética , Virulência , Yersinia/genética , Yersinia/imunologia , Yersinia/patogenicidade
16.
PLoS One ; 10(8): e0133298, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26275291

RESUMO

Microbial pathogens and host immune cells each initiate events following their interaction in an attempt to drive the outcome to their respective advantage. Here we show that the bacterial pathogen Yersinia pseudotuberculosis sustains itself on the surface of a macrophage by forming acidic fluid-accessible compartments that are partially bounded by the host cell plasma membrane. These Yersinia-containing acidic compartments (YACs) are bereft of the early endosomal marker EEA1 and the lysosomal antigen LAMP1 and readily form on primary macrophages as well as macrophage-like cell lines. YAC formation requires the presence of the Yersinia virulence plasmid which encodes a type III secretion system. Unexpectedly, we found that the initial formation of YACs did not require translocation of the type III effectors into the host cell cytosol; however, the duration of YACs was markedly greater in infections using translocation-competent Y. pseudotuberculosis strains as well as strains expressing the effector YopJ. Furthermore, it was in this translocation- and YopJ-dependent phase of infection that the acidic environment was critical for Y. pseudotuberculosis survival during its interaction with macrophages. Our findings indicate that during its extracellular phase of infection Y. pseudotuberculosis initiates and then, by a separate mechanism, stabilizes the formation of a highly intricate structure on the surface of the macrophage that is disengaged from the endocytic pathway.


Assuntos
Macrófagos/metabolismo , Macrófagos/microbiologia , Yersinia pseudotuberculosis/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Camundongos , Infecções por Yersinia pseudotuberculosis/metabolismo
17.
Immunol Res ; 57(1-3): 237-45, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24198067

RESUMO

Yersinia pestis, the etiologic agent of plague, utilizes a type III secretion system (T3SS) to subvert the defenses of its mammalian hosts. T3SSs are complex nanomachines that allow bacterial pathogens to directly inject effector proteins into eukaryotic cells. The Y. pestis T3SS is not expressed during transit through the flea vector, but T3SS gene expression is rapidly thermoinduced upon entry into a mammalian host. Assembly of the T3S apparatus is a highly coordinated process that requires the homo- and hetero-oligomerization over 20 Yersinia secretion (Ysc) proteins, several assembly intermediates and the T3S process to complete the assembly of the rod and external needle structures. The activation of effector secretion is controlled by the YopN/TyeA/SycN/YscB complex, YscF and LcrG in response to extracellular calcium and/or contact with a eukaryotic cell. Cell contact triggers the T3S process including the secretion and assembly of a pore-forming translocon complex that facilitates the translocation of effector proteins, termed Yersinia outer proteins (Yops), across the eukaryotic membrane. Within the host cell, the Yop effector proteins function to inhibit bacterial phagocytosis and to suppress the production of pro-inflammatory cytokines.


Assuntos
Sistemas de Secreção Bacterianos/fisiologia , Yersinia pestis/fisiologia , Animais , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Peste/imunologia , Peste/metabolismo , Peste/microbiologia , Ligação Proteica , Transporte Proteico , Transdução de Sinais
18.
PLoS One ; 8(7): e68754, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874749

RESUMO

Here we show that cells lacking the heme-regulated inhibitor (HRI) are highly resistant to infection by bacterial pathogens. By examining the infection process in wild-type and HRI null cells, we found that HRI is required for pathogens to execute their virulence-associated cellular activities. Specifically, unlike wild-type cells, HRI null cells infected with the gram-negative bacterial pathogen Yersinia are essentially impervious to the cytoskeleton-damaging effects of the Yop virulence factors. This effect is due to reduced functioning of the Yersinia type 3 secretion (T3S) system which injects virulence factors directly into the host cell cytosol. Reduced T3S activity is also observed in HRI null cells infected with the bacterial pathogen Chlamydia which results in a dramatic reduction in its intracellular proliferation. We go on to show that a HRI-mediated process plays a central role in the cellular infection cycle of the Gram-positive pathogen Listeria. For this pathogen, HRI is required for the post-invasion trafficking of the bacterium to the infected host cytosol. Thus by depriving Listeria of its intracellular niche, there is a highly reduced proliferation of Listeria in HRI null cells. We provide evidence that these infection-associated functions of HRI (an eIF2α kinase) are independent of its activity as a regulator of protein synthesis. This is the first report of a host factor whose absence interferes with the function of T3S secretion and cytosolic access by pathogens and makes HRI an excellent target for inhibitors due to its broad virulence-associated activities.


Assuntos
Bactérias/patogenicidade , Interações Hospedeiro-Patógeno , Fatores de Virulência/fisiologia , eIF-2 Quinase/fisiologia , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Células Cultivadas , Chlamydia trachomatis/patogenicidade , Resistência à Doença/genética , Feminino , Técnicas de Silenciamento de Genes , Células HeLa , Interações Hospedeiro-Patógeno/genética , Humanos , Listeria monocytogenes/patogenicidade , Masculino , Camundongos , Yersinia pseudotuberculosis/patogenicidade
19.
J Exp Med ; 208(13): 2705-16, 2011 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-22124111

RESUMO

Toll-like receptor 4 (TLR4), which signals through the adapter molecules myeloid differentiation factor 88 (MyD88) and toll/interleukin 1 receptor domain-containing adapter inducing IFN-ß (TRIF), is required for protection against Gram-negative bacteria. TRIF is known to be important in TLR3-mediated antiviral signaling, but the role of TRIF signaling against Gram-negative enteropathogens is currently unknown. We show that TRIF signaling is indispensable for establishing innate protective immunity against Gram-negative Yersinia enterocolitica. Infection of wild-type mice rapidly induced both IFN-ß and IFN-γ in the mesenteric lymph nodes. In contrast, TRIF-deficient mice were defective in these IFN responses and showed impaired phagocytosis in regional macrophages, resulting in greater bacterial dissemination and mortality. TRIF signaling may be universally important for protection against Gram-negative pathogens, as TRIF-deficient macrophages were also impaired in killing both Salmonella and Escherichia coli in vitro. The mechanism of TRIF-mediated protective immunity appears to be orchestrated by macrophage-induced IFN-ß and NK cell production of IFN-γ. Sequential induction of IFN-ß and IFN-γ leads to amplification of macrophage bactericidal activity sufficient to eliminate the invading pathogens at the intestinal interface. Our results demonstrate a previously unknown role of TRIF in host resistance to Gram-negative enteropathogens, which may lead to effective strategies for combating enteric infections.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Imunidade Inata/fisiologia , Células Matadoras Naturais/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Infecções por Bactérias Gram-Negativas/genética , Interferon beta/genética , Interferon beta/imunologia , Interferon gama/genética , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA