Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Leukoc Biol ; 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38416405

RESUMO

Lymph node stromal cells (LNSC) are an often overlooked component of the immune system, but play a crucial role in maintaining tissue homeostasis and orchestrating immune responses. Our understanding of the functions these cells serve in the context of bacterial infections remains limited. We previously showed that Listeria monocytogenes, a facultative intracellular foodborne bacterial pathogen, must replicate within an as-yet-unidentified cell type in the mesenteric lymph node (MLN) to spread systemically. Here, we show that L. monocytogenes could invade, escape from the vacuole, replicate exponentially, and induce a type I IFN response in the cytosol of two LNSC populations infected in vitro, fibroblastic reticular cells (FRC) and blood endothelial cells (BEC). Infected FRC and BEC also produced a significant chemokine and pro-inflammatory cytokine response after in vitro infection. Flow cytometric analysis confirmed that GFP+  L. monocytogenes were associated with a small percentage of MLN stromal cells in vivo following foodborne infection of mice. Using fluorescent microscopy, we showed that these cell-associated bacteria were intracellular L. monocytogenes and the number of infected FRC and BEC changed over the course of a three-day infection in mice. Ex vivo culturing of these infected LNSC populations revealed viable, replicating bacteria that grew on agar plates. These results highlight the unexplored potential of FRC and BEC to serve as suitable growth niches for L. monocytogenes during foodborne infection and to contribute to the pro-inflammatory environment within the MLN that promotes clearance of listeriosis.

2.
Infect Immun ; 91(9): e0025123, 2023 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-37594272

RESUMO

Apolipoprotein E (ApoE) is a lipid transport protein that is hypothesized to suppress proinflammatory cytokine production, particularly after stimulation with Toll-like receptor (TLR) ligands such as lipopolysaccharide (LPS). Studies using transgenic ApoE human replacement mice (APOE) expressing one of three different allelic variants suggest that there is a hierarchy in terms of responsiveness to proinflammatory stimuli such as APOE4/E4 > APOE3/E3 > APOE2/E2. In this study, we test the hypothesis that APOE genotype can also predict susceptibility to infection with the facultative intracellular gram-positive bacterium Listeria monocytogenes. We found that bone-marrow-derived macrophages isolated from aged APOE4/E4 mice expressed elevated levels of nitric oxide synthase 2 and were highly resistant to in vitro infection with L. monocytogenes compared to APOE3/E3 and APOE2/E2 mice. However, we did not find statistically significant differences in cytokine or chemokine output from either macrophages or whole splenocytes isolated from APOE2/E2, APOE3/E3, or APOE4/E4 mice following L. monocytogenes infection. In vivo, overall susceptibility to foodborne listeriosis also did not differ by APOE genotype in either young (2 mo old) or aged (15 mo old) C57BL/6 mice. However, we observed a sex-dependent susceptibility to infection in aged APOE2/E2 male mice and a sex-dependent resistance to infection in aged APOE4/E4 male mice that was not present in female mice. Thus, these results suggest that APOE genotype does not play an important role in innate resistance to infection with L. monocytogenes but may be linked to sex-dependent changes that occur during immune senescence.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Feminino , Humanos , Masculino , Camundongos , Apolipoproteína E2 , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Citocinas , Genótipo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
Infect Immun ; 91(4): e0006423, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-36916918

RESUMO

The mesenteric lymph nodes (MLN) function as a barrier to systemic spread for both commensal and pathogenic bacteria in the gut. Listeria monocytogenes, a facultative intracellular foodborne pathogen, readily overcomes this barrier and spreads into the bloodstream, causing life-threatening systemic infections. We show here that intracellular replication protected L. monocytogenes from clearance by monocytes and neutrophils and promoted colonization of the small intestine-draining MLN (sMLN) but was not required for dissemination to the colon-draining MLN (cMLN). Intestinal tissue had enough free lipoate to support LplA2-dependent extracellular growth of L. monocytogenes, but exogenous lipoate in the MLN was severely limited, and so the bacteria could replicate only inside cells, where they used LplA1 to scavenge lipoate from host peptides. When foodborne infection was manipulated to allow ΔlplA1 L. monocytogenes to colonize the MLN to the same extent as wild-type bacteria, the mutant was still never recovered in the spleen or liver of any animal. We found that intracellular replication in the MLN promoted actin-based motility and cell-to-cell spread of L. monocytogenes and that rapid efficient exit from the MLN was actA dependent. We conclude that intracellular replication of L. monocytogenes in intestinal tissues is not essential and serves primarily to amplify bacterial burdens above a critical threshold needed to efficiently colonize the cMLN. In contrast, intracellular replication in the MLN is absolutely required for further systemic spread and serves primarily to promote ActA-mediated cell-to-cell spread.


Assuntos
Listeria monocytogenes , Listeriose , Animais , Listeriose/microbiologia , Proteínas de Bactérias/genética , Fígado/patologia , Linfonodos/microbiologia
4.
Microbiol Resour Announc ; 10(18)2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-33958419

RESUMO

We report the whole-genome sequence of Listeria monocytogenes UKVDL9 and an edited draft genome sequence of L. monocytogenes 2010L-2198. Both are neurotropic lineage III strains; UKVDL9 was isolated from a sheep brain, and 2010L-2198 was isolated from a human subject with rhombencephalitis.

5.
mSphere ; 5(5)2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32938704

RESUMO

Listeria monocytogenes is thought to colonize the brain using one of three mechanisms: direct invasion of the blood-brain barrier, transportation across the barrier by infected monocytes, and axonal migration to the brain stem. The first two pathways seem to occur following unrestricted bacterial growth in the blood and thus have been linked to immunocompromise. In contrast, cell-to-cell spread within nerves is thought to be mediated by a particular subset of neurotropic L. monocytogenes strains. In this study, we used a mouse model of foodborne transmission to evaluate the neurotropism of several L. monocytogenes isolates. Two strains preferentially colonized the brain stems of BALB/cByJ mice 5 days postinfection and were not detectable in blood at that time point. In contrast, infection with other strains resulted in robust systemic infection of the viscera but no dissemination to the brain. Both neurotropic strains (L2010-2198, a human rhombencephalitis isolate, and UKVDL9, a sheep brain isolate) typed as phylogenetic lineage III, the least characterized group of L. monocytogenes Neither of these strains encodes InlF, an internalin-like protein that was recently shown to promote invasion of the blood-brain barrier. Acute neurologic deficits were observed in mice infected with the neurotropic strains, and milder symptoms persisted for up to 16 days in some animals. These results demonstrate that neurotropic L. monocytogenes strains are not restricted to any one particular lineage and suggest that the foodborne mouse model of listeriosis can be used to investigate the pathogenic mechanisms that allow L. monocytogenes to invade the brain stem.IMPORTANCE Progress in understanding the two naturally occurring central nervous system (CNS) manifestations of listeriosis (meningitis/meningoencephalitis and rhombencephalitis) has been limited by the lack of small animal models that can readily distinguish between these distinct infections. We report here that certain neurotropic strains of Listeria monocytogenes can spread to the brains of young otherwise healthy mice and cause neurological deficits without causing a fatal bacteremia. The novel strains described here fall within phylogenetic lineage III, a small collection of L. monocytogenes isolates that have not been well characterized to date. The animal model reported here mimics many features of human rhombencephalitis and will be useful for studying the mechanisms that allow L. monocytogenes to disseminate to the brain stem following natural foodborne transmission.


Assuntos
Encéfalo/microbiologia , Listeria monocytogenes/patogenicidade , Listeriose/sangue , Tropismo Viral , Animais , Encéfalo/patologia , Sistema Nervoso Central/microbiologia , Modelos Animais de Doenças , Feminino , Humanos , Encefalite Infecciosa/microbiologia , Listeria monocytogenes/isolamento & purificação , Listeriose/microbiologia , Listeriose/transmissão , Camundongos , Camundongos Endogâmicos BALB C , Filogenia , Ovinos , Virulência
6.
Curr Protoc Immunol ; 130(1): e102, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32710703

RESUMO

Listeria monocytogenes is a foodborne pathogen that causes serious, often deadly, systemic disease in susceptible individuals such as neonates and the elderly. These facultative intracellular bacteria have been an invaluable tool in immunology research for more than three decades. Intravenous (i.v.) injection is the most commonly used transmission route in mice, but oral models of infection have also been developed in recent years, and these may be more appropriate for many studies. This article includes detailed instructions for use of either foodborne or i.v. inoculation of mice and discusses the rationale for choosing either model. Additionally, a protocol is provided for enrichment of neutrophils and monocytes from the infected liver in a manner that allows for determination of bacterial burden while still providing sufficient cells for use in flow cytometric analysis or in vitro assays. © 2020 Wiley Periodicals LLC. Basic Protocol 1: Foodborne L. monocytogenes infection Support Protocol 1: Preparing L. monocytogenes for foodborne infection Basic Protocol 2: Intravenous L. monocytogenes infection Support Protocol 2: Preparing L. monocytogenes for intravenous infection Basic Protocol 3: Enrichment of non-parenchymal cells from the infected liver.


Assuntos
Listeria monocytogenes/fisiologia , Listeriose/etiologia , Listeriose/patologia , Fígado/microbiologia , Fígado/patologia , Monócitos/patologia , Neutrófilos/patologia , Animais , Biomarcadores , Biópsia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Doenças Transmitidas por Alimentos/microbiologia , Humanos , Imunofenotipagem , Listeriose/metabolismo , Listeriose/transmissão , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Especificidade da Espécie
7.
Proc Natl Acad Sci U S A ; 116(52): 26892-26899, 2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31818955

RESUMO

Mineral-respiring bacteria use a process called extracellular electron transfer to route their respiratory electron transport chain to insoluble electron acceptors on the exterior of the cell. We recently characterized a flavin-based extracellular electron transfer system that is present in the foodborne pathogen Listeria monocytogenes, as well as many other Gram-positive bacteria, and which highlights a more generalized role for extracellular electron transfer in microbial metabolism. Here we identify a family of putative extracellular reductases that possess a conserved posttranslational flavinylation modification. Phylogenetic analyses suggest that divergent flavinylated extracellular reductase subfamilies possess distinct and often unidentified substrate specificities. We show that flavinylation of a member of the fumarate reductase subfamily allows this enzyme to receive electrons from the extracellular electron transfer system and support L. monocytogenes growth. We demonstrate that this represents a generalizable mechanism by finding that a L. monocytogenes strain engineered to express a flavinylated extracellular urocanate reductase uses urocanate by a related mechanism and to a similar effect. These studies thus identify an enzyme family that exploits a modular flavin-based electron transfer strategy to reduce distinct extracellular substrates and support a multifunctional view of the role of extracellular electron transfer activities in microbial physiology.

8.
Microbiol Spectr ; 7(3)2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31124430

RESUMO

It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Animais , Vacinas Bacterianas/imunologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Humoral , Memória Imunológica , Camundongos , Linfócitos T/imunologia , Vacinas Atenuadas/imunologia
9.
J Immunol ; 202(12): 3474-3482, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31061007

RESUMO

PGE2 is a lipid-signaling molecule with complex roles in both homeostasis and inflammation. Depending on the cellular context, PGE2 may also suppress certain immune responses. In this study, we tested whether PGE2 could inhibit bacterial killing by polymorphonuclear neutrophils (PMN) using a mouse model of foodborne listeriosis. We found that PGE2 pretreatment decreased the ability of PMN harvested from the bone marrow of either BALB/cByJ or C57BL/6J mice to kill Listeria monocytogenes in vitro. PGE2 treatment slowed the migration of PMN toward the chemoattractant leukotriene B4, decreased uptake of L. monocytogenes by PMN, and inhibited the respiratory burst of PMN compared with vehicle-treated cells. When immune cells were isolated from the livers of infected mice and tested directly ex vivo for the presence of PGE2, BALB/cByJ cells produced significantly more than C57BL/6J cells. Together, these data suggest that robust PGE2 production can suppress PMN effector functions, leading to decreased bacterial killing, which may contribute to the innate susceptibility of BALB/cByJ mice to infection with the facultative intracellular bacterial pathogen L. monocytogenes.


Assuntos
Dinoprostona/metabolismo , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2/genética , Explosão Respiratória
10.
Infect Immun ; 86(4)2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29426040

RESUMO

Inbred mouse strains differ in their susceptibility to infection with the facultative intracellular bacterium Listeria monocytogenes, largely due to delayed or deficient innate immune responses. Previous antibody depletion studies suggested that neutrophils (polymorphonuclear leukocytes [PMN]) were particularly important for clearance in the liver, but the ability of PMN from susceptible and resistant mice to directly kill L. monocytogenes has not been examined. In this study, we showed that PMN infiltrated the livers of BALB/c/By/J (BALB/c) and C57BL/6 (B6) mice in similar numbers and that both cell types readily migrated toward leukotriene B4 in an in vitro chemotaxis assay. However, CFU burdens in the liver were significantly higher in BALB/c mice than in other strains, suggesting that PMN in the BALB/c liver might not be able to clear L. monocytogenes as efficiently as B6 PMN. Unprimed PMN harvested from either BALB/c or B6 bone marrow killed L. monocytogenes directly ex vivo, and pretreatment with autologous serum significantly enhanced killing efficiency for both. L. monocytogenes were internalized within 10 min and rapidly triggered intracellular production of reactive oxygen species in a dose-dependent manner. However, PMN from gp91phox-deficient mice also readily killed L. monocytogenes, which suggested that nonoxidative killing mechanisms may be sufficient for bacterial clearance. Together, these results indicate that there is not an intrinsic defect in the ability of PMN from susceptible BALB/c mice to kill L. monocytogenes and further suggest that if PMN function is impaired in BALB/c mice, it is likely due to locally produced modulating factors present in the liver during infection.


Assuntos
Resistência à Doença/imunologia , Suscetibilidade a Doenças/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose/imunologia , Animais , Biomarcadores , Imunidade Inata , Listeriose/metabolismo , Fígado/imunologia , Fígado/metabolismo , Fígado/microbiologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Viabilidade Microbiana/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória/imunologia , Especificidade da Espécie
11.
Pathogens ; 7(1)2018 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-29361677

RESUMO

Listeria monocytogenes is one of several enteric microbes that is acquired orally, invades the gastric mucosa, and then disseminates to peripheral tissues to cause systemic disease in humans. Intravenous (i.v.) inoculation of mice with L. monocytogenes has been the most widely-used small animal model of listeriosis over the past few decades. The infection is highly reproducible and has been invaluable in deciphering mechanisms of adaptive immunity in vivo, particularly CD8⁺ T cell responses to intracellular pathogens. However, the i.v. model completely bypasses the gut phase of the infection. Recent advances in generating both humanized mice and murinized bacteria, as well as the development of a foodborne route of transmission has reignited interest in studying oral models of listeriosis. In this review, we analyze previously published reports to highlight both the similarities and differences in tissue colonization and host response to infection using either oral or i.v. inoculation.

12.
Sci Rep ; 7(1): 17821, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29259308

RESUMO

The human pathogen L. monocytogenes and the animal pathogen L. ivanovii, together with four other species isolated from symptom-free animals, form the "Listeria sensu stricto" clade. The members of the second clade, "Listeria sensu lato", are believed to be solely environmental bacteria without the ability to colonize mammalian hosts. To identify novel determinants that contribute to infection by L. monocytogenes, the causative agent of the foodborne disease listeriosis, we performed a genome comparison of the two clades and found 151 candidate genes that are conserved in the Listeria sensu stricto species. Two factors were investigated further in vitro and in vivo. A mutant lacking an ATP-binding cassette transporter exhibited defective adhesion and invasion of human Caco-2 cells. Using a mouse model of foodborne L. monocytogenes infection, a reduced number of the mutant strain compared to the parental strain was observed in the small intestine and the liver. Another mutant with a defective 1,2-propanediol degradation pathway showed reduced persistence in the stool of infected mice, suggesting a role of 1,2-propanediol as a carbon and energy source of listeriae during infection. These findings reveal the relevance of novel factors for the colonization process of L. monocytogenes.


Assuntos
Listeria monocytogenes/genética , Listeriose/microbiologia , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Células CACO-2 , Linhagem Celular Tumoral , Feminino , Doenças Transmitidas por Alimentos/genética , Doenças Transmitidas por Alimentos/microbiologia , Humanos , Listeriose/genética , Camundongos , Camundongos Endogâmicos BALB C , Virulência/genética
13.
J Immunol ; 199(11): 3789-3797, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29055001

RESUMO

Recent fate-mapping studies and gene-expression profiles suggest that commonly used protocols to generate bone marrow-derived cultured dendritic cells yield a heterogeneous mixture, including some CD11chi cells that may not have a bona fide counterpart in vivo. In this study, we provide further evidence of the discordance between ex vivo-isolated and in vitro-cultured CD11c+ cells by analyzing an additional phenotype, the ability to support cytosolic growth of the facultative intracellular bacterial pathogen Listeria monocytogenes Two days after foodborne infection of mice with GFP-expressing L. monocytogenes, a small percentage of CD103neg and CD103+ conventional dendritic cells (cDC) in the intestinal lamina propria and mesenteric lymph nodes were GFP+ However, in vitro infection of the same subsets of cells harvested from naive mice resulted in inefficient invasion by the bacteria (<0.1% of the inoculum). The few intracellular bacteria detected survived for only a few hours. In contrast, cultured CD103negCD11c+ cells induced by GM-CSF readily supported exponential growth of L. monocytogenes Flt3 ligand-induced cultures yielded CD103+CD11c+ cells that more closely resembled cDC, with only a modest level of L. monocytogenes replication. For both culture protocols, the longer the cells were maintained in vitro, the more readily they supported intracellular growth. The results of this study suggest that cDC are not a niche for intracellular growth of L. monocytogenes during intestinal infection of mice.


Assuntos
Medula Óssea/imunologia , Células Dendríticas/imunologia , Trato Gastrointestinal/imunologia , Listeria monocytogenes/fisiologia , Listeriose/imunologia , Animais , Antígenos CD/metabolismo , Medula Óssea/microbiologia , Antígenos CD11/metabolismo , Processos de Crescimento Celular , Células Cultivadas , Replicação do DNA , DNA Bacteriano/genética , Células Dendríticas/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Imunofenotipagem , Cadeias alfa de Integrinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Tirosina Quinase 3 Semelhante a fms/metabolismo
14.
J Immunol ; 198(7): 2796-2804, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28213502

RESUMO

After foodborne transmission of the facultative intracellular bacterial pathogen Listeria monocytogenes, most of the bacterial burden in the gut is extracellular. However, we previously demonstrated that intracellular replication in an as yet unidentified cell type was essential for dissemination and systemic spread of L. monocytogenes In this article, we show that the vast majority of cell-associated L. monocytogenes in the gut were adhered to Ly6Chi monocytes, a cell type that inefficiently internalized L. monocytogenes With bone marrow-derived in vitro cultures, high multiplicity of infection or the use of opsonized bacteria enhanced uptake of L. monocytogenes in CD64- monocytes, but very few bacteria reached the cell cytosol. Surprisingly, monocytes that had upregulated CD64 expression in transition toward becoming macrophages fully supported intracellular growth of L. monocytogenes In contrast, inflammatory monocytes that had increased CD64 expression in the bone marrow of BALB/c/By/J mice prior to L. monocytogenes exposure in the gut did not support L. monocytogenes growth. Thus, contrary to the perception that L. monocytogenes can infect virtually all cell types, neither naive nor inflammatory Ly6Chi monocytes served as a productive intracellular growth niche for L. monocytogenes. These results have broad implications for innate immune recognition of L. monocytogenes in the gut and highlight the need for additional studies on the interaction of extracellular, adherent L. monocytogenes with the unique subsets of myeloid-derived inflammatory cells that infiltrate sites of infection.


Assuntos
Intestinos/imunologia , Listeriose/imunologia , Monócitos/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Intestinos/microbiologia , Listeria monocytogenes/imunologia , Listeria monocytogenes/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Virulência
15.
J Immunol ; 196(7): 3109-16, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26895837

RESUMO

Type I IFN (IFN-α/ß) is thought to enhance growth of the foodborne intracellular pathogen Listeria monocytogenes by promoting mechanisms that dampen innate immunity to infection. However, the type I IFN response has been studied primarily using methods that bypass the stomach and, therefore, fail to replicate the natural course of L. monocytogenes infection. In this study, we compared i.v. and foodborne transmission of L. monocytogenes in mice lacking the common type I IFN receptor (IFNAR1(-/-)). Contrary to what was observed using i.v. infection, IFNAR1(-/-) and wild-type mice had similar bacterial burdens in the liver and spleen following foodborne infection. Splenocytes from wild-type mice infected i.v. produced significantly more IFN-ß than did those infected by the foodborne route. Consequently, the immunosuppressive effects of type I IFN signaling, which included T cell death, increased IL-10 secretion, and repression of neutrophil recruitment to the spleen, were all observed following i.v. but not foodborne transmission of L. monocytogenes. Type I IFN was also previously shown to cause a loss of responsiveness to IFN-γ through downregulation of the IFN-γ receptor α-chain on macrophages and dendritic cells. However, we detected a decrease in surface expression of IFN-γ receptor α-chain even in the absence of IFN-α/ß signaling, suggesting that in vivo, this infection-induced phenotype is not type I IFN-dependent. These results highlight the importance of using the natural route of infection for studies of host-pathogen interactions and suggest that the detrimental effects of IFN-α/ß signaling on the innate immune response to L. monocytogenes may be an artifact of the i.v. infection model.


Assuntos
Suscetibilidade a Doenças , Interferon Tipo I/genética , Listeria monocytogenes/imunologia , Listeriose/genética , Listeriose/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Interferon Tipo I/metabolismo , Interferon beta/biossíntese , Listeriose/metabolismo , Depleção Linfocítica , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Transdução de Sinais , Baço/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
16.
Infect Immun ; 83(8): 3146-56, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26015479

RESUMO

Listeria monocytogenes is a highly adaptive bacterium that replicates as a free-living saprophyte in the environment as well as a facultative intracellular pathogen that causes invasive foodborne infections. The intracellular life cycle of L. monocytogenes is considered to be its primary virulence determinant during mammalian infection; however, the proportion of L. monocytogenes that is intracellular in vivo has not been studied extensively. In this report, we demonstrate that the majority of wild-type (strain EGDe) and mouse-adapted (InlA(m)-expressing) L. monocytogenes recovered from the mesenteric lymph nodes (MLN) was extracellular within the first few days after foodborne infection. In addition, significantly lower burdens of L. monocytogenes were recovered from the colon, spleen, and liver of gentamicin-treated mice than of control mice. This led us to investigate whether intracellular replication of L. monocytogenes was essential during the intestinal phase of infection. We found that lipoate protein ligase-deficient L. monocytogenes (ΔlplA1) mutants, which display impaired intracellular growth, were able to colonize the colon but did not persist efficiently and had a significant defect in spreading to the MLN, spleen, and liver. Together, these data indicate that the majority of the L. monocytogenes burden in the gastrointestinal tract is extracellular, but the small proportion of intracellular L. monocytogenes is essential for dissemination to the MLN and systemic organs.


Assuntos
Doenças Transmitidas por Alimentos/microbiologia , Intestinos/microbiologia , Listeria monocytogenes/crescimento & desenvolvimento , Listeriose/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Humanos , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Fígado/microbiologia , Linfonodos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/microbiologia
17.
PLoS Pathog ; 10(8): e1004301, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25101646

RESUMO

We characterized key components and major targets of the c-di-GMP signaling pathways in the foodborne pathogen Listeria monocytogenes, identified a new c-di-GMP-inducible exopolysaccharide responsible for motility inhibition, cell aggregation, and enhanced tolerance to disinfectants and desiccation, and provided first insights into the role of c-di-GMP signaling in listerial virulence. Genome-wide genetic and biochemical analyses of c-di-GMP signaling pathways revealed that L. monocytogenes has three GGDEF domain proteins, DgcA (Lmo1911), DgcB (Lmo1912) and DgcC (Lmo2174), that possess diguanylate cyclase activity, and three EAL domain proteins, PdeB (Lmo0131), PdeC (Lmo1914) and PdeD (Lmo0111), that possess c-di-GMP phosphodiesterase activity. Deletion of all phosphodiesterase genes (ΔpdeB/C/D) or expression of a heterologous diguanylate cyclase stimulated production of a previously unknown exopolysaccharide. The synthesis of this exopolysaccharide was attributed to the pssA-E (lmo0527-0531) gene cluster. The last gene of the cluster encodes the fourth listerial GGDEF domain protein, PssE, that functions as an I-site c-di-GMP receptor essential for exopolysaccharide synthesis. The c-di-GMP-inducible exopolysaccharide causes cell aggregation in minimal medium and impairs bacterial migration in semi-solid agar, however, it does not promote biofilm formation on abiotic surfaces. The exopolysaccharide also greatly enhances bacterial tolerance to commonly used disinfectants as well as desiccation, which may contribute to survival of L. monocytogenes on contaminated food products and in food-processing facilities. The exopolysaccharide and another, as yet unknown c-di-GMP-dependent target, drastically decrease listerial invasiveness in enterocytes in vitro, and lower pathogen load in the liver and gallbladder of mice infected via an oral route, which suggests that elevated c-di-GMP levels play an overall negative role in listerial virulence.


Assuntos
Proteínas de Bactérias/metabolismo , GMP Cíclico/análogos & derivados , Regulação Bacteriana da Expressão Gênica/fisiologia , Listeria monocytogenes/patogenicidade , Listeriose/metabolismo , Animais , Proteínas de Bactérias/genética , Cromatografia Líquida de Alta Pressão , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Proteínas de Escherichia coli/metabolismo , Feminino , Listeria monocytogenes/genética , Listeriose/genética , Camundongos , Camundongos Endogâmicos BALB C , Fósforo-Oxigênio Liases/metabolismo , Transdução de Sinais/fisiologia , Virulência/fisiologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-24575393

RESUMO

Listeria monocytogenes has been recognized as a food borne pathogen in humans since the 1980s, but we still understand very little about oral transmission of L. monocytogenes or the host factors that determine susceptibility to gastrointestinal infection, due to the lack of an appropriate small animal model of oral listeriosis. Early feeding trials suggested that many animals were highly resistant to oral infection, and the more reproducible intravenous or intraperitoneal routes of inoculation soon came to be favored. There are a fair number of previously published studies using an oral infection route, but the work varies widely in terms of bacterial strain choice, the methods used for oral transmission, and various manipulations used to enhance infectivity. This mini review summarizes the published literature using oral routes of L. monocytogenes infection and highlights recent technological advances that make oral infection a more attractive model system.


Assuntos
Modelos Animais de Doenças , Listeria monocytogenes/fisiologia , Listeriose/transmissão , Animais , Humanos , Listeria monocytogenes/isolamento & purificação , Listeriose/microbiologia
19.
J Vis Exp ; (75): e50381, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23685758

RESUMO

L. monocytogenes are facultative intracellular bacterial pathogens that cause food borne infections in humans. Very little is known about the gastrointestinal phase of listeriosis due to the lack of a small animal model that closely mimics human disease. This paper describes a novel mouse model for oral transmission of L. monocytogenes. Using this model, mice fed L. monocytogenes-contaminated bread have a discrete phase of gastrointestinal infection, followed by varying degrees of systemic spread in susceptible (BALB/c/By/J) or resistant (C57BL/6) mouse strains. During the later stages of the infection, dissemination to the gall bladder and brain is observed. The food borne model of listeriosis is highly reproducible, does not require specialized skills, and can be used with a wide variety of bacterial isolates and laboratory mouse strains. As such, it is the ideal model to study both virulence strategies used by L. monocytogenes to promote intestinal colonization, as well as the host response to invasive food borne bacterial infection.


Assuntos
Microbiologia de Alimentos , Listeria monocytogenes , Listeriose/transmissão , Animais , Feminino , Intestinos/microbiologia , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota
20.
Curr Protoc Microbiol ; 31: 9B.2.1-9B.2.7, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24510292

RESUMO

This unit describes general procedures for the lab cultivation and storage of the Gram-positive facultative intracellular bacterium Listeria monocytogenes. The basic protocols are relevant for a wide scope of applications including microbial genetics and both in vitro and in vivo infection studies. Commonly used L. monocytogenes strains, serotypes, and growth parameters are also discussed.


Assuntos
Técnicas Bacteriológicas/métodos , Listeria monocytogenes/crescimento & desenvolvimento , Preservação Biológica/métodos , Listeria monocytogenes/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...