Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.116
Filter
1.
Food Res Int ; 186: 114312, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38729688

ABSTRACT

Listeria monocytogenes exhibits varying levels of pathogenicity when entering the host through contaminated food. However, little is known regarding the stress response and environmental tolerance mechanism of different virulence strains to host gastrointestinal (GI) stimuli. This study analyzed the differences in the survival and genes of stress responses among two strains of L. monocytogenes 10403S (serotype 1/2a, highly virulent strain) and M7 (serotype 4a, low-virulence strain) during simulated gastrointestinal digestion. The results indicated that L. monocytogenes 10403S showed greater acid and bile salt tolerance than L. monocytogenes M7, with higher survival rates and less cell deformation and cell membrane permeability during the in vitro digestion. KEGG analysis of the transcriptomes indicated that L. monocytogenes 10403S displayed significant activity in amino acid metabolism, such as glutamate and arginine, associated with acid tolerance. Additionally, L. monocytogenes 10403S demonstrated a higher efficacy in promoting activities that preserve bacterial cell membrane integrity and facilitate flagellar protein synthesis. These findings will contribute valuable practical insights into the tolerance distinctions among different virulence strains of L. monocytogenes in the GI environment.


Subject(s)
Food Microbiology , Gastrointestinal Tract , Listeria monocytogenes , Meat Products , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Meat Products/microbiology , Virulence , Gastrointestinal Tract/microbiology , Bile Acids and Salts/metabolism , Digestion , Food Contamination , Microbial Viability , Cell Membrane Permeability
2.
Cell Syst ; 15(5): 425-444.e9, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38703772

ABSTRACT

The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.


Subject(s)
Placenta , Single-Cell Analysis , Humans , Female , Pregnancy , Placenta/microbiology , Placenta/immunology , Single-Cell Analysis/methods , Plasmodium falciparum , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/physiology , Toxoplasma/pathogenicity , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Toxoplasmosis/immunology , Toxoplasmosis/metabolism , Inflammation
3.
mBio ; 14(5): e0138523, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37706879

ABSTRACT

Folic acid and its derivatives are required for the synthesis of purines, pyrimidines, and some amino acids. Antifolate antibiotics that target the folic acid metabolism pathway are commonly used for the treatment of listeriosis caused by the intracellular pathogen Listeria monocytogenes (Lm). In recent work in mBio, Feng et al. sought to understand the role of folic acid metabolism in Lm virulence (Y. Feng, S. Chang, D. A. Portnoy, 2023, mBio https://doi.org/10.1128/mbio.01074-23). The authors discovered that N-formylmethionine, an amino acid utilized by bacteria to initiate protein synthesis, is crucial for Lm intracellular growth and pathogenesis. Surprisingly, purines and thymidine were found to be dispensable for Lm infection. Together these results demonstrate that while Lm can obtain many essential nutrients from the host cytosol, including purines and most amino acids, it requires N-formylmethionine biosynthesis to properly regulate translation initiation during infection.


Subject(s)
Listeria monocytogenes , Listeriosis , Humans , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Amino Acids/metabolism , Folic Acid/metabolism , Purines
4.
Can Vet J ; 64(4): 363-366, 2023 04.
Article in English | MEDLINE | ID: mdl-37008639

ABSTRACT

Clinical disease caused by infection with Listeria monocytogenes is rare in adult horses, and there is a paucity of ante-mortem clinicopathologic changes for this species reported in the literature. Confirmatory diagnosis is difficult and often requires post-mortem sampling of the brainstem. This report details a case of meningoencephalitis caused by Listeria monocytogenes in an adult American quarter horse gelding presenting with central neurologic signs. Pre-mortem analysis of the cerebrospinal fluid revealed a mononuclear, primarily lymphocytic, pleocytosis, which is a reported finding in other species with listeriosis. Post-mortem histopathologic changes of the brainstem were characteristic of listeriosis, and infection was confirmed with immunohistochemical labeling and bacterial culture. Key clinical message: Listeriosis should be included as a differential diagnosis in neurologic horses with mononuclear pleocytosis identified on cerebrospinal fluid analysis.


Pléocytose mononucléaire et méningo-encéphalite causées par Listeria monocytogenes chez un cheval adulte. La maladie clinique causée par une infection à L. monocytogenes est rare chez les chevaux adultes, et il y a peu de changements clinico-pathologiques ante-mortem rapportés dans la littérature pour cette espèce. Le diagnostic de confirmation est difficile et nécessite souvent un prélèvement post-mortem du tronc cérébral. Ce rapport détaille un cas de méningo-encéphalite causée par L. monocytogenes chez un hongre quarter horse américain adulte présentant des signes neurologiques centraux. L'analyse pré-mortem du liquide céphalo-rachidien a révélé une pléocytose mononucléaire, principalement lymphocytaire, qui est une trouvaille rapportée chez d'autres espèces atteintes de listériose. Les modifications histopathologiques post-mortem du tronc cérébral étaient caractéristiques de la listériose et l'infection a été confirmée par un marquage immunohistochimique et une culture bactérienne.Message clinique clé :La listériose doit être incluse comme diagnostic différentiel chez les chevaux avec signes neurologiques présentant une pléocytose mononucléaire identifiée lors de l'analyse du liquide céphalo-rachidien.(Traduit par Dr Serge Messier).


Subject(s)
Horse Diseases , Listeriosis , Meningoencephalitis , Animals , Male , Diagnosis, Differential , Horse Diseases/diagnosis , Horse Diseases/microbiology , Horses , Leukocytosis/diagnosis , Leukocytosis/veterinary , Listeria monocytogenes/pathogenicity , Listeriosis/diagnosis , Listeriosis/veterinary , Meningoencephalitis/diagnosis , Meningoencephalitis/microbiology , Meningoencephalitis/veterinary , Cerebrospinal Fluid/cytology
5.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 39(4): 289-294, 2023 Apr.
Article in Chinese | MEDLINE | ID: mdl-37087545

ABSTRACT

Objective To investigate the effect of long intergenic non-coding RNA COX2 (lincRNA-COX2) on apoptosis and polarization of Listeria monocytogenes (Lm)-infected RAW264.7 cells. Methods RAW264.7 cells were cultured and divided into control group (uninfected cells), Lm infection group, negative control of small interfering RNA (si-NC) group, si-NC and Lm infection group, small interfering RNA of lincRNA-COX2 (si-lincRNA-COX2) group, si-lincRNA-COX2 and Lm infection group. RAW264.7 cells were infected with MOI=10 Lm for 6 hours, and then the inhibition efficiency of siRNA transfection was detected by fluorescence microscope and quantitative real-time PCR (qRT-PCR). The expression levels of cleaved-caspase-3(c-caspase-3), caspase-3, B-cell lymphoma-2 (Bcl2), Bcl2 associated X protein (BAX), arginase 1 (Arg1), inducible nitric oxide synthase (iNOS) were detected by Western blot analysis. Results c-caspase-3/caspase-3, BAX/Bcl2 and iNOS were significantly up-regulated, while the level of Arg1 was down-regulated in Lm-infected RAW264.7 cells compared with control group. LincRNA-COX2 knockdown inhibited the increase of protein levels for BAX/Bcl2, c-caspase-3/caspase-3 and iNOS in Lm-infected RAW264.7 cells, while the level of Arg1 in Lm-infected RAW264.7 cells was up-regulated. Conclusion Knockdown of lincRNA-COX2 can inhibit cell apoptosis and suppress the macrophage polarization into M1 type in Lm-infected RAW264.7 cells.


Subject(s)
Cyclooxygenase 2 , Listeria monocytogenes , Macrophages , RNA, Long Noncoding , Apoptosis/genetics , bcl-2-Associated X Protein/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Macrophages/metabolism , Macrophages/microbiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , Animals , Mice
6.
Int J Mol Sci ; 24(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36902108

ABSTRACT

We explored the antimicrobial activity of sertraline on Listeria monocytogenes and further investigated the effects of sertraline on biofilm formation and the virulence gene expression of L. monocytogenes. The minimum inhibitory concentration and minimum bactericidal concentration for sertraline against L. monocytogenes were in the range of 16-32 µg/mL and 64 µg/mL, respectively. Sertraline-dependent damage of the cell membrane and a decrease in intracellular ATP and pHin in L. monocytogenes were observed. In addition, sertraline reduced the biofilm formation efficiency of the L. monocytogenes strains. Importantly, low concentrations (0.1 µg/mL and 1 µg/mL) of sertraline significantly down-regulated the expression levels of various L. monocytogens virulence genes (prfA, actA, degU, flaA, sigB, ltrC and sufS). These results collectively suggest a role of sertraline for the control of L. monocytogenes in the food industry.


Subject(s)
Anti-Infective Agents , Bacterial Proteins , Listeria monocytogenes , Sertraline , Virulence Factors , Anti-Infective Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/drug effects , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Sertraline/pharmacology , Virulence/drug effects , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-981867

ABSTRACT

Objective To investigate the effect of long intergenic non-coding RNA COX2 (lincRNA-COX2) on apoptosis and polarization of Listeria monocytogenes (Lm)-infected RAW264.7 cells. Methods RAW264.7 cells were cultured and divided into control group (uninfected cells), Lm infection group, negative control of small interfering RNA (si-NC) group, si-NC and Lm infection group, small interfering RNA of lincRNA-COX2 (si-lincRNA-COX2) group, si-lincRNA-COX2 and Lm infection group. RAW264.7 cells were infected with MOI=10 Lm for 6 hours, and then the inhibition efficiency of siRNA transfection was detected by fluorescence microscope and quantitative real-time PCR (qRT-PCR). The expression levels of cleaved-caspase-3(c-caspase-3), caspase-3, B-cell lymphoma-2 (Bcl2), Bcl2 associated X protein (BAX), arginase 1 (Arg1), inducible nitric oxide synthase (iNOS) were detected by Western blot analysis. Results c-caspase-3/caspase-3, BAX/Bcl2 and iNOS were significantly up-regulated, while the level of Arg1 was down-regulated in Lm-infected RAW264.7 cells compared with control group. LincRNA-COX2 knockdown inhibited the increase of protein levels for BAX/Bcl2, c-caspase-3/caspase-3 and iNOS in Lm-infected RAW264.7 cells, while the level of Arg1 in Lm-infected RAW264.7 cells was up-regulated. Conclusion Knockdown of lincRNA-COX2 can inhibit cell apoptosis and suppress the macrophage polarization into M1 type in Lm-infected RAW264.7 cells.


Subject(s)
Animals , Mice , Apoptosis/genetics , bcl-2-Associated X Protein/metabolism , Caspase 3/metabolism , Cyclooxygenase 2/metabolism , Listeria monocytogenes/pathogenicity , Macrophages/microbiology , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics
8.
PLoS One ; 17(9): e0274005, 2022.
Article in English | MEDLINE | ID: mdl-36054213

ABSTRACT

Listeria monocytogenes is a ubiquitous opportunistic foodborne pathogen capable of survival in various adverse environmental conditions. Pathogenesis of L. monocytogenes is tightly controlled by a complex regulatory network of transcriptional regulators that are necessary for survival and adaptations to harsh environmental conditions both inside and outside host cells. Among these regulatory pathways are members of the DeoR-family transcriptional regulators that are known to play a regulatory role in sugar metabolism. In this study, we deciphered the role of FruR, a DeoR family protein, which is a fructose operon transcriptional repressor protein, in L. monocytogenes pathogenesis and growth. Following intravenous (IV) inoculation in mice, a mutant strain with deletion of fruR exhibited a significant reduction in bacterial burden in liver and spleen tissues compared to the parent strain. Further, the ΔfruR strain had a defect in cell-to-cell spread in L2 fibroblast monolayers. Constitutive activation of PrfA, a pleiotropic activator of L. monocytogenes virulence factors, did not restore virulence to the ΔfruR strain, suggesting that the attenuation was not a result of impaired PrfA activation. Transcriptome analysis revealed that FruR functions as a positive regulator for genes encoding enzymes involved in the pentose phosphate pathway (PPP) and as a repressor for genes encoding enzymes in the glycolysis pathway. These results suggested that FruR may function to facilitate NADPH regeneration, which is necessary for full protection from oxidative stress. Interestingly, deletion of fruR increased sensitivity of L. monocytogenes to H2O2, confirming a role for FruR in survival of L. monocytogenes during oxidative stress. Using anti-mouse neutrophil/monocyte monoclonal antibody RB6-8C5 (RB6) in an in vivo infection model, we found that FruR has a specific function in protecting L. monocytogenes from neutrophil/monocyte-mediated killing. Overall, this work clarifies the role of FruR in controlling L. monocytogenes carbon flow between glycolysis and PPP for NADPH homeostasis, which provides a new mechanism allowing metabolic adaptation of L. monocytogenes to oxidative stress.


Subject(s)
Listeria monocytogenes , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Hydrogen Peroxide/metabolism , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Mice , Peptide Termination Factors/metabolism , Regulon , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence
9.
Proc Natl Acad Sci U S A ; 119(13): e2122173119, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35316134

ABSTRACT

Flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD) are essential riboflavin-derived cofactors involved in a myriad of redox reactions across all forms of life. Nevertheless, the basis of flavin acquisition strategies by riboflavin auxotrophic pathogens remains poorly defined. In this study, we examined how the facultative intracellular pathogen Listeria monocytogenes, a riboflavin auxotroph, acquires flavins during infection. A L. monocytogenes mutant lacking the putative riboflavin transporter (RibU) was completely avirulent in mice but had no detectable growth defect in nutrient-rich media. However, unlike wild type, the RibU mutant was unable to grow in defined media supplemented with FMN or FAD or to replicate in macrophages starved for riboflavin. Consistent with RibU functioning to scavenge FMN and FAD inside host cells, a mutant unable to convert riboflavin to FMN or FAD retained virulence and grew in cultured macrophages and in spleens and livers of infected mice. However, this FMN- and FAD-requiring strain was unable to grow in the gallbladder or intestines, where L. monocytogenes normally grows extracellularly, suggesting that these sites do not contain sufficient flavin cofactors to promote replication. Thus, by deleting genes required to synthesize FMN and FAD, we converted L. monocytogenes from a facultative to an obligate intracellular pathogen. Collectively, these data indicate that L. monocytogenes requires riboflavin to grow extracellularly in vivo but scavenges FMN and FAD to grow in host cells.


Subject(s)
Bacterial Proteins , Flavin Mononucleotide , Flavin-Adenine Dinucleotide , Listeria monocytogenes , Membrane Transport Proteins , Riboflavin , Bacterial Proteins/metabolism , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Listeria monocytogenes/growth & development , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Membrane Transport Proteins/metabolism , Riboflavin/metabolism
10.
Nature ; 603(7903): 900-906, 2022 03.
Article in English | MEDLINE | ID: mdl-35296858

ABSTRACT

Infections of the central nervous system are among the most serious infections1,2, but the mechanisms by which pathogens access the brain remain poorly understood. The model microorganism Listeria monocytogenes (Lm) is a major foodborne pathogen that causes neurolisteriosis, one of the deadliest infections of the central nervous system3,4. Although immunosuppression is a well-established host risk factor for neurolisteriosis3,5, little is known about the bacterial factors that underlie the neuroinvasion of Lm. Here we develop a clinically relevant experimental model of neurolisteriosis, using hypervirulent neuroinvasive strains6 inoculated in a humanized mouse model of infection7, and we show that the bacterial surface protein InlB protects infected monocytes from Fas-mediated cell death by CD8+ T cells in a manner that depends on c-Met, PI3 kinase and FLIP. This blockade of specific anti-Lm cellular immune killing lengthens the lifespan of infected monocytes, and thereby favours the transfer of Lm from infected monocytes to the brain. The intracellular niche that is created by InlB-mediated cell-autonomous immune resistance also promotes Lm faecal shedding, which accounts for the selection of InlB as a core virulence gene of Lm. We have uncovered a specific mechanism by which a bacterial pathogen confers an increased lifespan to the cells it infects by rendering them resistant to cell-mediated immunity. This promotes the persistence of Lm within the host, its dissemination to the central nervous system and its transmission.


Subject(s)
Central Nervous System Diseases , Listeria monocytogenes , Listeriosis , Animals , Bacterial Proteins/metabolism , CD8-Positive T-Lymphocytes/metabolism , Central Nervous System Diseases/microbiology , Disease Models, Animal , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Mice , Monocytes , Virulence
11.
Cell Rep ; 38(8): 110414, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35196496

ABSTRACT

Inflammasome activation exacerbates infectious disease caused by pathogens such as Listeria monocytogenes, Staphylococcus aureus, and severe acute respiratory syndrome coronavirus 2. Although these pathogens activate host inflammasomes to regulate pathogen expansion, the mechanisms by which pathogen toxins contribute to inflammasome activation remain poorly understood. Here we show that activation of inflammasomes by Listeria infection is promoted by amino acid residue T223 of listeriolysin O (LLO) independently of its pore-forming activity. LLO T223 is critical for phosphorylation of the inflammasome adaptor ASC at amino acid residue Y144 through Lyn-Syk signaling, which is essential for ASC oligomerization. Notably, a Listeria mutant expressing LLO T223A is impaired in inducing ASC phosphorylation and inflammasome activation. Furthermore, the virulence of LLO T223A mutant is markedly attenuated in vivo due to impaired ability to activate the inflammasome. Our results reveal a function of a pathogen toxin that exacerbates infection by promoting phosphorylation of ASC.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Inflammasomes/metabolism , Listeria monocytogenes/pathogenicity , Signal Transduction , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/deficiency , CARD Signaling Adaptor Proteins/genetics , Gene Editing , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Interleukin-18/metabolism , Listeria monocytogenes/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Phosphorylation , Syk Kinase/genetics , Syk Kinase/metabolism , Virulence , src-Family Kinases/genetics , src-Family Kinases/metabolism
12.
PLoS Pathog ; 18(1): e1010166, 2022 01.
Article in English | MEDLINE | ID: mdl-35007292

ABSTRACT

A hallmark of Listeria (L.) monocytogenes pathogenesis is bacterial escape from maturing entry vacuoles, which is required for rapid bacterial replication in the host cell cytoplasm and cell-to-cell spread. The bacterial transcriptional activator PrfA controls expression of key virulence factors that enable exploitation of this intracellular niche. The transcriptional activity of PrfA within infected host cells is controlled by allosteric coactivation. Inhibitory occupation of the coactivator site has been shown to impair PrfA functions, but consequences of PrfA inhibition for L. monocytogenes infection and pathogenesis are unknown. Here we report the crystal structure of PrfA with a small molecule inhibitor occupying the coactivator site at 2.0 Å resolution. Using molecular imaging and infection studies in macrophages, we demonstrate that PrfA inhibition prevents the vacuolar escape of L. monocytogenes and enables extensive bacterial replication inside spacious vacuoles. In contrast to previously described spacious Listeria-containing vacuoles, which have been implicated in supporting chronic infection, PrfA inhibition facilitated progressive clearance of intracellular L. monocytogenes from spacious vacuoles through lysosomal degradation. Thus, inhibitory occupation of the PrfA coactivator site facilitates formation of a transient intravacuolar L. monocytogenes replication niche that licenses macrophages to effectively eliminate intracellular bacteria. Our findings encourage further exploration of PrfA as a potential target for antimicrobials and highlight that intra-vacuolar residence of L. monocytogenes in macrophages is not inevitably tied to bacterial persistence.


Subject(s)
Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Macrophages/microbiology , Vacuoles/microbiology , Virulence/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL
13.
PLoS Pathog ; 17(12): e1010173, 2021 12.
Article in English | MEDLINE | ID: mdl-34929015

ABSTRACT

For many intracellular bacterial pathogens manipulating host cell survival is essential for maintaining their replicative niche, and is a common strategy used to promote infection. The bacterial pathogen Listeria monocytogenes is well known to hijack host machinery for its own benefit, such as targeting the host histone H3 for modification by SIRT2. However, by what means this modification benefits infection, as well as the molecular players involved, were unknown. Here we show that SIRT2 activity supports Listeria intracellular survival by maintaining genome integrity and host cell viability. This protective effect is dependent on H3K18 deacetylation, which safeguards the host genome by counteracting infection-induced DNA damage. Mechanistically, infection causes SIRT2 to interact with the nucleic acid binding protein TDP-43 and localise to genomic R-loops, where H3K18 deacetylation occurs. This work highlights novel functions of TDP-43 and R-loops during bacterial infection and identifies the mechanism through which L. monocytogenes co-opts SIRT2 to allow efficient infection.


Subject(s)
Histones/metabolism , Host-Pathogen Interactions/physiology , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Sirtuin 3/metabolism , Animals , Cell Survival/physiology , Humans , Listeria monocytogenes/metabolism
14.
Cell Rep ; 37(5): 109956, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731605

ABSTRACT

Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Liver/immunology , Malaria/immunology , Plasmodium berghei/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/parasitology , Disease Models, Animal , Female , Host-Parasite Interactions , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/blood , Listeriosis/immunology , Listeriosis/microbiology , Liver/metabolism , Liver/microbiology , Liver/parasitology , Lymphocyte Function-Associated Antigen-1/metabolism , Malaria/blood , Malaria/parasitology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Parasite Load , Phagocytes/immunology , Phagocytes/metabolism , Phagocytes/microbiology , Phagocytes/parasitology , Plasmodium berghei/pathogenicity , Time Factors
15.
Nat Commun ; 12(1): 6826, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819495

ABSTRACT

Listeria genus comprises two pathogenic species, L. monocytogenes (Lm) and L. ivanovii, and non-pathogenic species. All can thrive as saprophytes, whereas only pathogenic species cause systemic infections. Identifying Listeria species' respective biotopes is critical to understand the ecological contribution of Listeria virulence. In order to investigate the prevalence and abundance of Listeria species in various sources, we retrieved and analyzed 16S rRNA datasets from MG-RAST metagenomic database. 26% of datasets contain Listeria sensu stricto sequences, and Lm is the most prevalent species, most abundant in soil and host-associated environments, including 5% of human stools. Lm is also detected in 10% of human stool samples from an independent cohort of 900 healthy asymptomatic donors. A specific microbiota signature is associated with Lm faecal carriage, both in humans and experimentally inoculated mice, in which it precedes Lm faecal carriage. These results indicate that Lm faecal carriage is common and depends on the gut microbiota, and suggest that Lm faecal carriage is a crucial yet overlooked consequence of its virulence.


Subject(s)
Carrier State/epidemiology , Gastrointestinal Microbiome/genetics , Listeria monocytogenes/isolation & purification , Animals , Carrier State/diagnosis , Carrier State/microbiology , DNA, Bacterial/isolation & purification , Datasets as Topic , Disease Models, Animal , Feces/microbiology , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Male , Metagenomics/statistics & numerical data , Mice , Phylogeny , RNA, Ribosomal, 16S/genetics , Virulence
16.
Int J Rheum Dis ; 24(11): 1427-1439, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34633142

ABSTRACT

AIM: To review the clinical features of systemic lupus erythematosus (SLE) complicated by central nervous system (CNS) infection due to Listeria monocytogenes. METHOD: A patient with SLE receiving high-dose glucocorticoids combined with cyclophosphamide who developed multiple brain abscesses due to Listeria infection is described. The case is compared with known cases in a literature review. RESULTS: A review of the literature showed that CNS infections are rare bacterial complications of SLE, but they can be a significant cause of mortality, especially those due to L. monocytogenes. The most significant risk factor for listerial meningitis is a prior history of receiving immunosuppressive therapy. At-risk patients should avoid unpasteurized milk and soft cheeses along with deli-style, ready-to-eat prepared meats, particularly poultry products. The case we report is the fifth SLE patient with multiple brain abscesses due to L. monocytogenes, and the first to be discharged with no sequelae. Timely and accurate identification and treatment of CNS infections and neuropsychiatric lupus are very important for favorable disease prognosis. CONCLUSION: Repeated blood culture is helpful for early diagnosis, and empirical anti-infective treatment that covers L. monocytogenes is recommended for SLE patients with risk factors when CNS infection occurs. A comprehensive assessment might be helpful to distinguish CNS infections from neuropsychiatric SLE. For severe infection, the dosage of steroids does not need to be reduced immediately but can be gradually adjusted based on the results of a comprehensive evaluation of the disease.


Subject(s)
Brain Abscess/microbiology , Cyclophosphamide/adverse effects , Glucocorticoids/adverse effects , Immunosuppressive Agents/adverse effects , Listeria monocytogenes/pathogenicity , Lupus Erythematosus, Systemic/drug therapy , Meningitis, Listeria/microbiology , Anti-Bacterial Agents/therapeutic use , Brain Abscess/diagnosis , Brain Abscess/drug therapy , Brain Abscess/immunology , Female , Humans , Immunocompromised Host , Listeria monocytogenes/immunology , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/immunology , Meningitis, Listeria/diagnosis , Meningitis, Listeria/drug therapy , Meningitis, Listeria/immunology , Middle Aged , Risk Factors , Treatment Outcome
17.
Nat Immunol ; 22(11): 1382-1390, 2021 11.
Article in English | MEDLINE | ID: mdl-34663978

ABSTRACT

Intergenerational inheritance of immune traits linked to epigenetic modifications has been demonstrated in plants and invertebrates. Here we provide evidence for transmission of trained immunity across generations to murine progeny that survived a sublethal systemic infection with Candida albicans or a zymosan challenge. The progeny of trained mice exhibited cellular, developmental, transcriptional and epigenetic changes associated with the bone marrow-resident myeloid effector and progenitor cell compartment. Moreover, the progeny of trained mice showed enhanced responsiveness to endotoxin challenge, alongside improved protection against systemic heterologous Escherichia coli and Listeria monocytogenes infections. Sperm DNA of parental male mice intravenously infected with the fungus C. albicans showed DNA methylation differences linked to immune gene loci. These results provide evidence for inheritance of trained immunity in mammals, enhancing protection against infections.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Heredity , Immunity, Innate/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Myeloid Cells/immunology , Animals , Candida albicans/pathogenicity , Candidiasis/genetics , Candidiasis/metabolism , Candidiasis/microbiology , Cells, Cultured , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Escherichia coli/pathogenicity , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Male , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/microbiology , Spermatozoa/immunology , Spermatozoa/metabolism , Transcription, Genetic
18.
Mol Microbiol ; 116(6): 1407-1419, 2021 12.
Article in English | MEDLINE | ID: mdl-34704304

ABSTRACT

Listeria monocytogenes is a food-borne bacterium that causes gastroenteritis, meningitis, or abortion. L. monocytogenes induces its internalization (entry) into human cells and either spreads laterally in tissues or transcytoses to traverse anatomical barriers. In this review, we discuss mechanisms by which five structurally related proteins of the "internalin" family of L. monocytogenes (InlA, InlB, InlC, InlF, and InlP) interact with distinct host receptors to promote infection of human cells and/or crossing of the intestinal, blood-brain, or placental barriers. We focus on recent results demonstrating that the internalin proteins InlA, InlB, and InlC exploit exocytic pathways to stimulate transcytosis, entry, or cell-to-cell spread, respectively. We also discuss evidence that InlA-mediated transcytosis contributes to traversal of the intestinal barrier, whereas InlF promotes entry into endothelial cells to breach the blood-brain barrier. InlB also facilitates the crossing of the blood-brain barrier, but does so by extending the longevity of infected monocytes that may subsequently act as a "Trojan horse" to transfer bacteria to the brain. InlA, InlB, and InlP each contribute to fetoplacental infection by targeting syncytiotrophoblast or cytotrophoblast layers of the placenta. This work highlights the diverse functions of internalins and the complex mechanisms by which these structurally related proteins contribute to disease.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Animals , Bacterial Proteins/genetics , Humans , Listeria monocytogenes/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Transcytosis
19.
FEBS Open Bio ; 11(12): 3262-3275, 2021 12.
Article in English | MEDLINE | ID: mdl-34709730

ABSTRACT

Bacterial phospholipases and sphingomyelinases are lipolytic esterases that are structurally and evolutionarily heterogeneous. These enzymes play crucial roles as virulence factors in several human and animal infectious diseases. Some bacterial phospholipases C (PLCs) have both phosphatidylcholinesterase and sphingomyelinase C activities. Among them, Listeria monocytogenes PlcB, Clostridium perfringens PLC, and Pseudomonas aeruginosa PlcH are the most deeply understood. In silico predictions of substrates docking with these three bacterial enzymes provide evidence that they interact with different substrates at the same active site. This review discusses structural aspects, substrate specificity, and the mechanism of action of those bacterial enzymes on target cells and animal infection models to shed light on their roles in pathogenesis.


Subject(s)
Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/physiology , Type C Phospholipases/metabolism , Type C Phospholipases/physiology , Animals , Clostridium perfringens/enzymology , Clostridium perfringens/pathogenicity , Humans , Listeria monocytogenes/enzymology , Listeria monocytogenes/pathogenicity , Phospholipases , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity , Type C Phospholipases/genetics
20.
PLoS Pathog ; 17(10): e1009881, 2021 10.
Article in English | MEDLINE | ID: mdl-34624065

ABSTRACT

Pathogenic bacteria rely on protein phosphorylation to adapt quickly to stress, including that imposed by the host during infection. Penicillin-binding protein and serine/threonine-associated (PASTA) kinases are signal transduction systems that sense cell wall integrity and modulate multiple facets of bacterial physiology in response to cell envelope stress. The PASTA kinase in the cytosolic pathogen Listeria monocytogenes, PrkA, is required for cell wall stress responses, cytosolic survival, and virulence, yet its substrates and downstream signaling pathways remain incompletely defined. We combined orthogonal phosphoproteomic and genetic analyses in the presence of a ß-lactam antibiotic to define PrkA phosphotargets and pathways modulated by PrkA. These analyses synergistically highlighted ReoM, which was recently identified as a PrkA target that influences peptidoglycan (PG) synthesis, as an important phosphosubstrate during cell wall stress. We find that deletion of reoM restores cell wall stress sensitivities and cytosolic survival defects of a ΔprkA mutant to nearly wild-type levels. While a ΔprkA mutant is defective for PG synthesis during cell wall stress, a double ΔreoM ΔprkA mutant synthesizes PG at rates similar to wild type. In a mouse model of systemic listeriosis, deletion of reoM in a ΔprkA background almost fully restored virulence to wild-type levels. However, loss of reoM alone also resulted in attenuated virulence, suggesting ReoM is critical at some points during pathogenesis. Finally, we demonstrate that the PASTA kinase/ReoM cell wall stress response pathway is conserved in a related pathogen, methicillin-resistant Staphylococcus aureus. Taken together, our phosphoproteomic analysis provides a comprehensive overview of the PASTA kinase targets of an important model pathogen and suggests that a critical role of PrkA in vivo is modulating PG synthesis through regulation of ReoM to facilitate cytosolic survival and virulence.


Subject(s)
Cell Wall/physiology , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Peptidoglycan/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Listeriosis/metabolism , Mice , Mice, Inbred C57BL , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...