Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.411
Filter
1.
Gut Microbes ; 16(1): 2369564, 2024.
Article in English | MEDLINE | ID: mdl-38979800

ABSTRACT

The tight and coordinated regulation of virulence gene expression is crucial to ensure the survival and persistence of bacterial pathogens in different contexts within their hosts. Considering this, bacteria do not express virulence factors homogenously in time and space, either due to their associated fitness cost or to their detrimental effect at specific infection stages. To efficiently infect and persist into their hosts, bacteria have thus to monitor environmental cues or chemical cell-to-cell signaling mechanisms that allow their transition from the external environment to the host, and therefore adjust gene expression levels, intrinsic biological activities, and appropriate behaviors. Listeria monocytogenes (Lm), a major Gram-positive facultative intracellular pathogen, stands out for its adaptability and capacity to thrive in a wide range of environments. Because of that, Lm presents itself as a significant concern in food safety and public health, that can lead to potentially life-threatening infections in humans. A deeper understanding of the intricate bacterial virulence mechanisms and the signals that control them provide valuable insights into the dynamic interplay between Lm and the host. Therefore, this review addresses the role of some crucial signals behind Lm pathogenic virulence mechanisms and explores how the ability to assimilate and interpret these signals is fundamental for pathogenesis, identifying potential targets for innovative antimicrobial strategies.


Subject(s)
Gene Expression Regulation, Bacterial , Listeria monocytogenes , Listeriosis , Virulence Factors , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/physiology , Humans , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Listeriosis/microbiology , Animals , Signal Transduction , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Host-Pathogen Interactions
2.
Front Cell Infect Microbiol ; 14: 1411124, 2024.
Article in English | MEDLINE | ID: mdl-39045131

ABSTRACT

Dissemination of food-borne L. monocytogenes in the host relies on internalin-mediated invasion, but the underlying invasion strategies remain elusive. Here we use live-cell microscopy to follow single cell interactions between individual human cells and L. monocytogenes and elucidate mechanisms associated with internalin B (InlB)-mediated invasion. We demonstrate that whilst a replicative invasion of nonphagocytic cells is a rare event even at high multiplicities of invasion, L. monocytogenes overcomes this by utilising a strategy relaying on PrfA-mediated ActA-based aggregation. We show that L. monocytogenes forms aggregates in extracellular host cell environment, which promote approximately 5-fold more host cell adhesions than the non-aggregating actA-ΔC mutant (which lacks the C-terminus coding region), with the adhering bacteria inducing 3-fold more intracellular invasions. Aggregation is associated with robust MET tyrosine kinase receptor clustering in the host cells, a hallmark of InlB-mediated invasion, something not observed with the actA-ΔC mutant. Finally, we show via RNA-seq analyses that aggregation involves a global adaptive response to host cell environment (including iron depletion), resulting in metabolic changes in L. monocytogenes and upregulation of the PrfA virulence regulon. Overall, our analyses provide new mechanistic insights into internalin-mediated host-pathogen interactions of L. monocytogenes.


Subject(s)
Bacterial Adhesion , Bacterial Proteins , Listeria monocytogenes , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/physiology , Listeria monocytogenes/metabolism , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Host-Pathogen Interactions , Listeriosis/microbiology , Peptide Termination Factors/metabolism , Peptide Termination Factors/genetics , Gene Expression Regulation, Bacterial , Virulence/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
3.
Nature ; 631(8021): 635-644, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961291

ABSTRACT

Innate immune pattern recognition receptors, such as the Toll-like receptors (TLRs), are key mediators of the immune response to infection and central to our understanding of health and disease1. After microbial detection, these receptors activate inflammatory signal transduction pathways that involve IκB kinases, mitogen-activated protein kinases, ubiquitin ligases and other adaptor proteins. The mechanisms that connect the proteins in the TLR pathways are poorly defined. To delineate TLR pathway activities, we engineered macrophages to enable microscopy and proteomic analysis of the endogenous myddosome constituent MyD88. We found that myddosomes form transient contacts with activated TLRs and that TLR-free myddosomes are dynamic in size, number and composition over the course of 24 h. Analysis using super-resolution microscopy revealed that, within most myddosomes, MyD88 forms barrel-like structures that function as scaffolds for effector protein recruitment. Proteomic analysis demonstrated that myddosomes contain proteins that act at all stages and regulate all effector responses of the TLR pathways, and genetic analysis defined the epistatic relationship between these effector modules. Myddosome assembly was evident in cells infected with Listeria monocytogenes, but these bacteria evaded myddosome assembly and TLR signalling during cell-to-cell spread. On the basis of these findings, we propose that the entire TLR signalling pathway is executed from within the myddosome.


Subject(s)
Macrophages , Signal Transduction , Toll-Like Receptors , Animals , Humans , Mice , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Myeloid Differentiation Factor 88/metabolism , Proteomics , Toll-Like Receptors/metabolism , Microscopy , Immunity, Innate
4.
BMC Vet Res ; 20(1): 297, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971767

ABSTRACT

BACKGROUND: Listeriosis is a global health threat to both animals and humans, especially in developing countries. This study was designed to isolate Listeria monocytogenes from faeces; environmental samples; and cow, sheep and goat milk, as well as human stool, to study its molecular characteristics and antibiotic sensitivity in the New Valley and Beheira Governorates, Egypt. The isolation and identification of L. monocytogenes were carried out using traditional culture and biochemical methods, followed by antibiography, genus confirmation of some isolates and detection and sequencing of InlB genes via PCR. RESULTS: Out of 2097 examined samples, the prevalence of L. monocytogenes was 13.4% in animals; the prevalence was 9.2%, 2.4%, 25.4%, 4%, 42.4%, and 6.4% in cattle faeces, cattle milk, sheep faeces, sheep milk, goat faeces, and goat milk, respectively. However, the prevalence of L. monocytogenes was 8.3% in human samples. Both animal and human isolates showed 100% resistance to trimethoprim-sulfamethoxazole, and the isolates showed the highest sensitivity to flumequine (100%), amikacin (99.2%), gentamicin (97.6%), and levofloxacin (94.6%). Multidrug resistance (MDR) was detected in 86.9% of the tested isolates. The 16 S rRNA and inlB genes were detected in 100% of the randomly selected L. monocytogenes isolates. Phylogenetic analysis of three isolates based on the inlB gene showed 100% identity between faecal, milk and human stool isolates. CONCLUSIONS: Faeces and milk are major sources of listeriosis, and the high degree of genetic similarity between animal and human isolates suggests the possibility of zoonotic circulation. The high prevalence of MDR L. monocytogenes in both animal and human samples could negatively impact the success of prevention and treatments for animal and human diseases, thereby imposing serious risks to public health.


Subject(s)
Anti-Bacterial Agents , Feces , Goats , Listeria monocytogenes , Listeriosis , Milk , Animals , Egypt/epidemiology , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Listeria monocytogenes/isolation & purification , Humans , Prevalence , Sheep , Anti-Bacterial Agents/pharmacology , Cattle , Feces/microbiology , Listeriosis/veterinary , Listeriosis/epidemiology , Listeriosis/microbiology , Milk/microbiology , Microbial Sensitivity Tests , Drug Resistance, Bacterial/genetics
5.
Ann Agric Environ Med ; 31(2): 311-314, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38940119

ABSTRACT

Listeria monocytogenes is a Gram-positive facultative anaerobic bacterium that is ubiquitous in the environment and can cause severe infections in immunocompromised individuals, pregnant women, and newborns. Listeriosis can manifest as meningitis, encephalitis, or sepsis, and its diagnosis requires a high index of suspicion. The case is reported of a rare presentation of rhombencephalitis by listeriosis in a 61-year-old male who initially suffered from subacute gastric disturbances and fever. Neurological consultation showed abnormal functions of cranial nerves and meningeal signs were observed. MRI revealed a poorly demarcated focus of approximately 45 × 16 × 15mm, indicating possible inflammatory processes, necessitating a lumbar puncture. Assessment of the CSF indicated infection with the bacterium- Listeria Monocytogenes, with the final diagnosis of Listeriosis encephalitis. Despite antibiotic therapy of Ceftazidine and Ampicillin, the patient's condition deteriorated, followed by death.


Subject(s)
Encephalitis , Listeria monocytogenes , Listeriosis , Humans , Male , Listeriosis/diagnosis , Listeriosis/drug therapy , Listeriosis/microbiology , Middle Aged , Fatal Outcome , Listeria monocytogenes/isolation & purification , Encephalitis/microbiology , Encephalitis/drug therapy , Encephalitis/diagnosis , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/administration & dosage , Rhombencephalon/microbiology
6.
J Bacteriol ; 206(7): e0003324, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38899896

ABSTRACT

Listeria monocytogenes is a foodborne bacterial pathogen that causes listeriosis. Positive regulatory factor A (PrfA) is a pleiotropic master activator of virulence genes of L. monocytogenes that becomes active upon the entry of the bacterium into the cytosol of infected cells. L. monocytogenes can survive and multiply at low temperatures; this is accomplished through the maintenance of appropriate membrane fluidity via branched-chain fatty acid (BCFA) synthesis. Branched-chain α-keto acid dehydrogenase (BKD), which is composed of four polypeptides encoded by lpd, bkdA1, bkdA2, and bkdB, is known to play a vital role in BCFA biosynthesis. Here, we constructed BKD-deficient Listeria strains by in-frame deletion of lpd, bkdA1, bkdA2, and bkdB genes. To determine the role in in vivo and in vitro, mouse model challenges, plaque assay in murine L2 fibroblast, and intracellular replication in J744A.1 macrophage were conducted. BKD-deficient strains exhibited defects in BCFA composition, virulence, and PrfA-regulon function within the host cells. Transcriptomics analysis revealed that the transcript level of the PrfA-regulon was lower in ΔbkdA1 strain than those in the wild-type. This study demonstrates that L. monocytogenes strains lacking BKD complex components were defective in PrfA-regulon function, and full activation of wild-type prfA may not occur within host cells in the absence of BKD. Further study will investigate the consequences of BKD deletion on PrfA function through altering BCFA catabolism.IMPORTANCEListeria monocytogenes is the causative agent of listeriosis, a disease with a high mortality rate. In this study, we have shown that the deletion of BKD can impact the function of PrfA and the PrfA-regulon. The production of virulence proteins within host cells is necessary for L. monocytogenes to promote its intracellular survival and is likely dependent on membrane integrity. We thus report a link between L. monocytogenes membrane integrity and the function of PrfA. This knowledge will increase our understanding of L. monocytogenes pathogenesis, which may provide insight into the development of antimicrobial agents.


Subject(s)
Bacterial Proteins , Listeria monocytogenes , Listeriosis , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Mice , Animals , Virulence , Listeriosis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fatty Acids/biosynthesis , Fatty Acids/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/metabolism , 3-Methyl-2-Oxobutanoate Dehydrogenase (Lipoamide)/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Female , Cell Line
7.
Appl Microbiol Biotechnol ; 108(1): 384, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896287

ABSTRACT

Bacteriocins have the potential to effectively improve food-borne infections or gastrointestinal diseases and hold promise as viable alternatives to antibiotics. This study aimed to explore the antibacterial activity of three bacteriocins (nisin, enterocin Gr17, and plantaricin RX-8) and their ability to attenuate intestinal barrier dysfunction and inflammatory responses induced by Listeria monocytogenes, respectively. Bacteriocins have shown excellent antibacterial activity against L. monocytogenes without causing any cytotoxicity. Bacteriocins inhibited the adhesion and invasion of L. monocytogenes on Caco-2 cells, lactate dehydrogenase (LDH), trans-epithelial electrical resistance (TEER), and cell migration showed that bacteriocin improved the permeability of Caco-2 cells. These results were attributed to the promotion of tight junction proteins (TJP) assembly, specifically zonula occludens-1 (ZO-1), occludin, and claudin-1. Furthermore, bacteriocins could alleviate inflammation by inhibiting the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways and reducing the secretion of interleukin-6 (IL-6), interleukin-1 ß (IL-1ß) and tumor necrosis factor α (TNF-α). Among three bacteriocins, plantaricin RX-8 showed the best antibacterial activity against L. monocytogenes and the most pronounced protective effect on the intestinal barrier due to its unique structure. Based on our findings, we hypothesized that bacteriocins may inhibit the adhesion and invasion of L. monocytogenes by competing adhesion sites. Moreover, they may further enhance intestinal barrier function by inhibiting the expression of L. monocytogenes virulence factors, increasing the expression of TJP and decreasing the secretion of inflammatory factors. Therefore, bacteriocins will hopefully be an effective alternative to antibiotics, and this study provides valuable insights into food safety concerns. KEY POINTS: • Bacteriocins show excellent antibacterial activity against L. monocytogenes • Bacteriocins improve intestinal barrier damage and inflammatory response • Plantaricin RX-8 has the best protective effect on Caco-2 cells damage.


Subject(s)
Anti-Bacterial Agents , Bacteriocins , Listeria monocytogenes , Listeria monocytogenes/drug effects , Bacteriocins/pharmacology , Humans , Caco-2 Cells , Anti-Bacterial Agents/pharmacology , Inflammation , NF-kappa B/metabolism , Bacterial Adhesion/drug effects , Tight Junction Proteins/metabolism , Cytokines/metabolism , Listeriosis/microbiology , Listeriosis/drug therapy , Cell Movement/drug effects
8.
Appl Environ Microbiol ; 90(6): e0213523, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38727222

ABSTRACT

Listeria monocytogenes, a prominent foodborne pathogen responsible for zoonotic infections, owes a significant portion of its virulence to the presence of the phospholipase PlcB. In this study, we performed an in-depth examination of the intricate relationship between L. monocytogenes PlcB and host cell mitochondria, unveiling a novel participant in bacterial survival: the mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA). Our investigation uncovered previously unexplored levels of interaction and colocalization between PCCA and PlcB within host cells, with particular emphasis on the amino acids 504-508 of PCCA, which play a pivotal role in this partnership. To assess the effect of PCCA expression on L. monocytogenes proliferation, PCCA expression levels were manipulated by siRNA-si-PCCA or pCMV-N-HA-PCCA plasmid transfection. Our findings demonstrated a clear inverse correlation between PCCA expression levels and the proliferation of L. monocytogenes. Furthermore, the effect of L. monocytogenes infection on PCCA expression was investigated by assessing PCCA mRNA and protein expression in HeLa cells infected with L. monocytogenes. These results indicate that L. monocytogenes infection did not significantly alter PCCA expression. These findings led us to propose that PCCA represents a novel participant in L. monocytogenes survival, and its abundance has a detrimental impact on bacterial proliferation. This suggests that L. monocytogenes may employ PlcB-PCCA interactions to maintain stable PCCA expression, representing a unique pro-survival strategy distinct from that of other intracellular bacterial pathogens. IMPORTANCE: Mitochondria represent attractive targets for pathogenic bacteria seeking to modulate host cellular processes to promote their survival and replication. Our current study has uncovered mitochondrial carboxylase propionyl-coenzyme A carboxylase (PCCA) as a novel host cell protein that interacts with L. monocytogenes PlcB. The results demonstrate that PCCA plays a negative regulatory role in L. monocytogenes infection, as heightened PCCA levels are associated with reduced bacterial survival and persistence. However, L. monocytogenes may exploit the PlcB-PCCA interaction to maintain stable PCCA expression and establish a favorable intracellular milieu for bacterial infection. Our findings shed new light on the intricate interplay between bacterial pathogens and host cell mitochondria, while also highlighting the potential of mitochondrial metabolic enzymes as antimicrobial agents.


Subject(s)
Bacterial Proteins , Listeria monocytogenes , Listeria monocytogenes/genetics , Listeria monocytogenes/enzymology , Humans , HeLa Cells , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mitochondria/metabolism , Listeriosis/microbiology , Microbial Viability
9.
Infect Immun ; 92(6): e0016224, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38752742

ABSTRACT

Ethanolamine (EA) affects the colonization and pathogenicity of certain human bacterial pathogens in the gastrointestinal tract. However, EA can also affect the intracellular survival and replication of host cell invasive bacteria such as Listeria monocytogenes (LMO) and Salmonella enterica serovar Typhimurium (S. Typhimurium). The EA utilization (eut) genes can be categorized as regulatory, enzymatic, or structural, and previous work in LMO showed that loss of genes encoding functions for the enzymatic breakdown of EA inhibited LMO intracellular replication. In this work, we sought to further characterize the role of EA utilization during LMO infection of host cells. Unlike what was previously observed for S. Typhimurium, in LMO, an EA regulator mutant (ΔeutV) was equally deficient in intracellular replication compared to an EA metabolism mutant (ΔeutB), and this was consistent across Caco-2, RAW 264.7, and THP-1 cell lines. The structural genes encode proteins that self-assemble into bacterial microcompartments (BMCs) that encase the enzymes necessary for EA metabolism. For the first time, native EUT BMCs were fluorescently tagged, and EUT BMC formation was observed in vitro and in vivo. Interestingly, BMC formation was observed in bacteria infecting Caco-2 cells, but not the macrophage cell lines. Finally, the cellular immune response of Caco-2 cells to infection with eut mutants was examined, and it was discovered that ΔeutB and ΔeutV mutants similarly elevated the expression of inflammatory cytokines. In conclusion, EA sensing and utilization during LMO intracellular infection are important for optimal LMO replication and immune evasion but are not always concomitant with BMC formation.IMPORTANCEListeria monocytogenes (LMO) is a bacterial pathogen that can cause severe disease in immunocompromised individuals when consumed in contaminated food. It can replicate inside of mammalian cells, escaping detection by the immune system. Therefore, understanding the features of this human pathogen that contribute to its infectiousness and intracellular lifestyle is important. In this work we demonstrate that genes encoding both regulators and enzymes of EA metabolism are important for optimal growth inside mammalian cells. Moreover, the formation of specialized compartments to enable EA metabolism were visualized by tagging with a fluorescent protein and found to form when LMO infects some mammalian cell types, but not others. Interestingly, the formation of the compartments was associated with features consistent with an early stage of the intracellular infection. By characterizing bacterial metabolic pathways that contribute to survival in host environments, we hope to positively impact knowledge and facilitate new treatment strategies.


Subject(s)
Ethanolamine , Listeria monocytogenes , Listeriosis , Listeria monocytogenes/metabolism , Listeria monocytogenes/growth & development , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Humans , Ethanolamine/metabolism , Mice , Animals , RAW 264.7 Cells , Caco-2 Cells , THP-1 Cells , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Macrophages/microbiology , Macrophages/metabolism
10.
BMC Infect Dis ; 24(1): 477, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720244

ABSTRACT

We report a very rare case of Listeria multiple brain abscesses manifested as delirium, which represented diagnostic and therapeutic challenges overcome only by the close cooperation between Infectious Diseases and Neuroradiology, without which a satisfactory outcome would not be achieved.An elderly man presented with confusion and drowsiness with a background of type-II diabetes mellitus. Although computed tomography of the brain only showed frontal lobe oedema, contrast magnetic resonance (MR) imaging showed numerous irregular rim-enhancing lesions containing central diffusion restriction, suggesting multiple pyogenic cerebral abscesses of unclear aetiology. Thereafter, Listeria monocytogenes was isolated from blood cultures, suggesting this as the causative organism. Deemed unsuitable for neurosurgical drainage, the patient received medical management with a protracted course of antibiotics. This case was extremely challenging, due to 1) the impossibility of source control, 2) the small number of effective antibiotics available to treat this condition, and 3) the inevitable antibiotic side-effects, derived from long-term exposure. A successful outcome was only possible thanks to strict close multidisciplinary follow up, requiring frequent MR imaging and a judicious antibiotic choice, including monitoring of their side-effects. Due to the rarity of this condition, there is lack of guidance on its management, hence the importance of multidisciplinary involvement with very close imaging and antibiotic monitoring.


Subject(s)
Anti-Bacterial Agents , Brain Abscess , Listeria monocytogenes , Listeriosis , Humans , Male , Brain Abscess/microbiology , Brain Abscess/drug therapy , Brain Abscess/diagnostic imaging , Listeriosis/drug therapy , Listeriosis/microbiology , Listeriosis/diagnosis , Anti-Bacterial Agents/therapeutic use , Listeria monocytogenes/isolation & purification , Aged , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Brain/diagnostic imaging , Brain/pathology , Brain/microbiology , Delirium/drug therapy
11.
Euro Surveill ; 29(19)2024 May.
Article in English | MEDLINE | ID: mdl-38726694

ABSTRACT

Listeria monocytogenes (Lm) is a bacterium widely distributed in the environment. Listeriosis is a severe disease associated with high hospitalisation and mortality rates. In April 2019, listeriosis was diagnosed in two hospital patients in Finland. We conducted a descriptive study to identify the source of the infection and defined a case as a person with a laboratory-confirmed Lm serogroup IIa sequence type (ST) 37. Six cases with Lm ST 37 were notified to the Finnish Infectious Diseases Registry between 2015 and 2019. Patient interviews and hospital menus were used to target traceback investigation of the implicated foods. In 2021 and 2022, similar Lm ST 37 was detected from samples of a ready-to-eat plant-based food product including fava beans. Inspections by the manufacturer and the local food control authority indicated that the food products were contaminated with Lm after pasteurisation. Our investigation highlights the importance that companies producing plant-based food are subject to similar controls as those producing food of animal origin. Hospital menus can be a useful source of information that is not dependent on patient recall.


Subject(s)
Disease Outbreaks , Food Microbiology , Listeria monocytogenes , Listeriosis , Humans , Listeria monocytogenes/isolation & purification , Listeria monocytogenes/genetics , Listeriosis/epidemiology , Listeriosis/microbiology , Finland/epidemiology , Female , Male , Foodborne Diseases/epidemiology , Foodborne Diseases/microbiology , Middle Aged , Aged , Food Contamination , Adult , Fabaceae/microbiology
12.
Front Biosci (Landmark Ed) ; 29(5): 176, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38812301

ABSTRACT

BACKGROUND: Listeria monocytogenes, a Gram-positive bacterium, is a prominent foodborne pathogen that causes listeriosis and poses substantial health hazards worldwide. The continuing risk of listeriosis outbreaks underlies the importance of designing an effective prevention strategy and developing a robust immune response by reverse vaccinology approaches. This study aimed to provide a critical approach for developing a potent multiepitope vaccine against this foodborne disease. METHODS: A chimeric peptide construct containing 5 B-cell epitopes, 16 major histocompatibility complex I (MHC-I) epitopes, and 18 MHC-II epitopes were used to create a subunit vaccination against L. monocytogenes. The vaccine safety was evaluated by several online methods, and molecular docking was performed using ClusPro to determine the binding affinity. Immune simulation was performed using the C-ImmSimm server to demonstrate the immune response. RESULTS: The results validated the antigenicity, non-allergenicity, and nontoxicity of the chimeric peptide construct, confirming its suitability as a subunit vaccine. Molecular docking showed a good score of 1276.5 and molecular dynamics simulations confirmed the construct's efficacy, demonstrating its promise as a good candidate for listeriosis prophylaxis. The population coverage was as high as 91.04% with a good immune response, indicating good antigen presentation with dendritic cells and production of memory cells. CONCLUSIONS: The findings of this study highlight the potential of the designed chimeric peptide construct as an effective subunit vaccine against Listeria, paving the way for future advances in preventive methods and vaccine design.


Subject(s)
Bacterial Vaccines , Computational Biology , Listeria monocytogenes , Listeriosis , Molecular Docking Simulation , Vaccines, Subunit , Listeria monocytogenes/immunology , Bacterial Vaccines/immunology , Vaccines, Subunit/immunology , Listeriosis/prevention & control , Listeriosis/immunology , Listeriosis/microbiology , Computational Biology/methods , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/chemistry , Humans , Epitopes/immunology , Molecular Dynamics Simulation , Animals , Foodborne Diseases/prevention & control , Foodborne Diseases/microbiology , Foodborne Diseases/immunology , Immunoinformatics
13.
Sci Adv ; 10(22): eadj1431, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38809979

ABSTRACT

Infusion of 13C-labeled metabolites provides a gold standard for understanding the metabolic processes used by T cells during immune responses in vivo. Through infusion of 13C-labeled metabolites (glucose, glutamine, and acetate) in Listeria monocytogenes-infected mice, we demonstrate that CD8 T effector (Teff) cells use metabolites for specific pathways during specific phases of activation. Highly proliferative early Teff cells in vivo shunt glucose primarily toward nucleotide synthesis and leverage glutamine anaplerosis in the tricarboxylic acid (TCA) cycle to support adenosine triphosphate and de novo pyrimidine synthesis. In addition, early Teff cells rely on glutamic-oxaloacetic transaminase 1 (Got1)-which regulates de novo aspartate synthesis-for effector cell expansion in vivo. CD8 Teff cells change fuel preference over the course of infection, switching from glutamine- to acetate-dependent TCA cycle metabolism late in infection. This study provides insights into the dynamics of Teff metabolism, illuminating distinct pathways of fuel consumption associated with CD8 Teff cell function in vivo.


Subject(s)
Acetates , CD8-Positive T-Lymphocytes , Carbon Isotopes , Glutamine , Glutamine/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Acetates/metabolism , Mice , Listeriosis/metabolism , Listeriosis/immunology , Listeriosis/microbiology , Listeria monocytogenes , Citric Acid Cycle , Glucose/metabolism , Mice, Inbred C57BL
14.
Microbiol Spectr ; 12(7): e0050924, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38809017

ABSTRACT

Bacterial genotyping through whole-genome sequencing plays a crucial role in disease surveillance and outbreak investigations in public health laboratories. This study assessed the effectiveness of Oxford Nanopore Technologies (ONT) sequencing in the genotyping of Listeria monocytogenes and Salmonella enterica serovar Enteritidis. Our results indicated that ONT sequences, generated with the R10.4.1 flow cell and basecalled using the Dorado 0.5.0 Super Accurate 4.3 model, exhibited comparable accuracy to Illumina sequences, effectively discriminating among bacterial strains from outbreaks. These findings suggest that ONT sequencing has the potential to be a promising tool for rapid whole-genome sequencing of bacterial pathogens in public health laboratories for epidemiological investigations. IMPORTANCE: This study unveils that Oxford Nanopore Technologies sequencing, by itself, holds the potential to serve as a whole-genome sequencing-based genotyping tool in public health laboratories, enabling routine subtyping of bacterial isolates for disease surveillance and outbreak investigations.


Subject(s)
Genome, Bacterial , Listeria monocytogenes , Nanopore Sequencing , Salmonella enteritidis , Whole Genome Sequencing , Listeria monocytogenes/genetics , Listeria monocytogenes/classification , Listeria monocytogenes/isolation & purification , Salmonella enteritidis/genetics , Salmonella enteritidis/classification , Salmonella enteritidis/isolation & purification , Whole Genome Sequencing/methods , Nanopore Sequencing/methods , Genome, Bacterial/genetics , Humans , Listeriosis/microbiology , Genotype , Disease Outbreaks , Genotyping Techniques/methods , Salmonella Infections/microbiology
15.
Anal Chem ; 96(21): 8543-8551, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38748432

ABSTRACT

In this study, the covalently fixed "end-on" orientation of a monoclonal Listeria monocytogenes antibody (mAb-Lis) to amino terminated oligo (ethylene glycol)-capped gold nanoparticles (NH2-TEG-AuNPs) was used to fabricate an in-house lateral flow strip (LFS), namely, the fixed "end-on" Lis-mAb-NH-TEG-AuNPs LFS. The aim was to evaluate the performance of the fixed "end-on" Lis-mAb-NH-TEG-AuNPs LFS in detecting L. monocytogenes. The proposed LFS enabled the sensitive detection of L. monocytogenes in 15 min with a visual limit of detection of 102 CFU/mL. Quantitative analysis indicated an LOD at 10 CFU/mL. The fixed "end-on" Lis-mAb-NH-TEG-AuNPs LFS showed no cross-reactivity with other pathogenic bacteria and practical performance across different food matrices, including human blood, milk, and mushroom samples. Furthermore, the clinical performance of the fixed "end-on" Lis-mAb-NH-TEG-AuNPs LFS for detecting L. monocytogenes was evaluated by using 12 clinical samples validated by the hemoculture method. It demonstrated excellent concordance with the reference methods, with no false-positive or false-negative results observed. Therefore, the fixed "end-on" Lis-mAb-NH-TEG-AuNPs LFS serves as a promising candidate for a point-of-care test (POCT), enabling the rapid, precise, and highly sensitive detection of L. monocytogenes in clinical samples and contaminated food.


Subject(s)
Antibodies, Monoclonal , Gold , Listeria monocytogenes , Metal Nanoparticles , Listeria monocytogenes/isolation & purification , Listeria monocytogenes/immunology , Gold/chemistry , Metal Nanoparticles/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/chemistry , Humans , Limit of Detection , Food Microbiology , Milk/microbiology , Milk/chemistry , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/immunology , Antibodies, Immobilized/chemistry , Antibodies, Immobilized/immunology , Animals , Listeriosis/microbiology , Listeriosis/diagnosis
16.
mBio ; 15(7): e0074324, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38809022

ABSTRACT

Pathogenic bacteria rely on secreted virulence factors to cause disease in susceptible hosts. However, in Gram-positive bacteria, the mechanisms underlying secreted protein activation and regulation post-membrane translocation remain largely unknown. Using proteomics, we identified several proteins that are dependent on the secreted chaperone PrsA2. We followed with phenotypic, biochemical, and biophysical assays and computational analyses to examine the regulation of a detected key secreted virulence factor, listeriolysin O (LLO), and its interaction with PrsA2 from the bacterial pathogen Listeria monocytogenes (Lm). Critical to Lm virulence is internalization by host cells and the subsequent action of the cholesterol-dependent pore-forming toxin, LLO, which enables bacterial escape from the host cell phagosome. Since Lm is a Gram-positive organism, the space between the cell membrane and wall is solvent exposed. Therefore, we hypothesized that the drop from neutral to acidic pH as the pathogen is internalized into a phagosome is critical to regulating the interaction of PrsA2 with LLO. Here, we demonstrate that PrsA2 directly interacts with LLO in a pH-dependent manner. We show that PrsA2 protects and sequesters LLO under neutral pH conditions where LLO can be observed to aggregate. In addition, we identify molecular features of PrsA2 that are required for interaction and ultimately the folding and activity of LLO. Moreover, protein-complex modeling suggests that PrsA2 interacts with LLO via its cholesterol-binding domain. These findings highlight a mechanism by which a Gram-positive secretion chaperone regulates the secretion, stability, and folding of a pore-forming toxin under conditions relevant to host cell infection. IMPORTANCE: Lm is a ubiquitous food-borne pathogen that can cause severe disease to vulnerable populations. During infection, Lm relies on a wide repertoire of secreted virulence factors including the LLO that enables the bacterium to invade the host and spread from cell to cell. After membrane translocation, secreted factors must become active in the challenging bacterial cell membrane-wall interface. However, the mechanisms required for secreted protein folding and function are largely unknown. Lm encodes a chaperone, PrsA2, that is critical for the activity of secreted factors. Here, we show that PrsA2 directly associates and protects the major Lm virulence factor, LLO, under conditions corresponding to the host cytosol, where LLO undergoes irreversible denaturation. Additionally, we identify molecular features of PrsA2 that enable its interaction with LLO. Together, our results suggest that Lm and perhaps other Gram-positive bacteria utilize secreted chaperones to regulate the activity of pore-forming toxins during infection.


Subject(s)
Bacterial Toxins , Heat-Shock Proteins , Hemolysin Proteins , Listeria monocytogenes , Listeriosis , Protein Folding , Hemolysin Proteins/metabolism , Hemolysin Proteins/genetics , Hemolysin Proteins/chemistry , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/chemistry , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeria monocytogenes/pathogenicity , Listeria monocytogenes/chemistry , Bacterial Toxins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/chemistry , Listeriosis/microbiology , Virulence Factors/metabolism , Virulence Factors/genetics , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/chemistry , Peptidylprolyl Isomerase/metabolism , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/chemistry , Hydrogen-Ion Concentration , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Protein Stability , Humans
17.
Lancet Infect Dis ; 24(7): 783-792, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608698

ABSTRACT

BACKGROUND: Listeriosis is a foodborne infection caused by Listeria monocytogenes. Three main forms of listeriosis are well characterised, but little is known about L monocytogenes-associated spontaneous bacterial peritonitis. We used data from the French national surveillance of listeriosis to perform a nationwide retrospective study. METHODS: All patients with L monocytogenes isolated by culture from a peritoneal fluid sample in France between April 1, 1993, and Dec 31, 2022, were included. Individuals for whom bacterial peritonitis was not confirmed and those who also had another type of invasive listeriosis were excluded. A standardised checklist was used to collect demographic, clinical, and biological data as well as antibiotic treatment and follow-up data. The primary outcome was to determine the characteristics of L monocytogenes-associated spontaneous bacterial peritonitis. We did descriptive analyses and assessed risk factors for 1-month mortality using an exploratory multivariable Cox model analysis. FINDINGS: Among the 8768 L monocytogenes cases reported, 208 (2%) were patients with L monocytogenes-associated spontaneous bacterial peritonitis. Mean age was 65 years (SD 13), 50 (24%) of 208 patients were female, and 158 (76%) were male (no data on race or ethnicity were available). 200 (98%) of 205 patients with L monocytogenes-associated spontaneous bacterial peritonitis with available data had immunosuppressive comorbidities, including cirrhosis (148 [74%] of 201 with available data), ongoing alcoholism (58 [62%] of 94), and ongoing neoplasia (60 [31%] of 195). Causes of ascites included cirrhosis (146 [70%] of 208), ongoing neoplasia (26 [13%]), end-stage heart failure (13 [6%]), and peritoneal dialysis (11 [5%]). Among those with available data, presentation was pauci-symptomatic and non-specific; only 67 (50%) of 135 patients presented with fever, 49 (37%) of 132 with abdominal pain, and 27 (21%) of 129 with diarrhoea. 61 (29%) of 208 patients were dead at 1 month, 92 (44%) were dead at 3 months, and 109 (52%) were dead at 6 months after diagnosis. Ongoing neoplasia (hazard ratio 2·42 [95% CI 1·05-5·56]; p=0·039), septic shock (8·03 [2·66-24·02]; p=0·0021), and high blood leukocyte count (1·05 [1·00-1·09]; p=0·045) were independently associated with 1-month mortality. INTERPRETATION: Despite the non-specific and mild presentation of L monocytogenes-associated spontaneous bacterial peritonitis, the outcome is poor and similar to that of neurolisteriosis, and so identification of L monocytogenes in ascitic fluid samples requires urgent parenteral amoxicillin-based treatment to avoid a fatal outcome. FUNDING: Institut Pasteur, Inserm, and French Public Health Agency. TRANSLATION: For the French translation of the abstract see Supplementary Materials section.


Subject(s)
Listeria monocytogenes , Listeriosis , Peritonitis , Humans , Male , Female , Peritonitis/microbiology , Peritonitis/mortality , Peritonitis/epidemiology , Peritonitis/drug therapy , Listeriosis/epidemiology , Listeriosis/mortality , Listeriosis/microbiology , Listeriosis/complications , France/epidemiology , Aged , Listeria monocytogenes/isolation & purification , Middle Aged , Retrospective Studies , Risk Factors , Aged, 80 and over , Anti-Bacterial Agents/therapeutic use , Adult
18.
Vet Microbiol ; 293: 110086, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615477

ABSTRACT

Listeriosis is a zoonotic disease caused by Listeria monocytogenes and Listeria ivanovii. The genus Listeria currently includes 27 recognized species and is found throughout the environment. The number of systematic studies on antimicrobial resistance in L. monocytogenes isolates from domestic farms using antimicrobial substances is limited. Importantly, dairy ruminant farms are reservoir of hypervirulent lineage I L. monocytogenes isolates, previously associated with human clinical cases. Considering that the classes of antibiotics used in food-producing domestic animals are frequently the same or closely related to those used in human medicine, studies about the impact of antibiotic use on the acquisition of antibiotic resistance in Listeria spp. in domestic animal farms are, therefore, of high importance. Here, susceptibility to 25 antibiotics was determined. Eighty-one animal-related, 35 food and 21 human pathogenic Listeria spp. isolates and 114 animal-related non-pathogenic Listeria spp. isolates were tested. Whole genome sequencing data was used for molecular characterization. Regarding L. monocytogenes, 2 strains from the clinical-associated linage I showed resistance to erythromycin, both related to dairy ruminants. Acquired resistance to one antibiotic was exhibited in 1.5% of L. monocytogenes isolates compared with 14% of non-pathogenic Listeria spp. isolates. Resistance to tetracycline (7.9%), doxycycline (7.9%), penicillin (4.4%), and ampicillin (4.4%) were the most frequently observed in non-pathogenic Listeria spp. While resistance to two or more antibiotics (5.6%) was most common in Listeria spp., isolates, resistance to one antibiotic was also observed (1.6%). The present results show that non-pathogenic Listeria spp. harbour antimicrobial resistance genes.


Subject(s)
Anti-Bacterial Agents , Listeria , Listeriosis , Microbial Sensitivity Tests , Animals , Listeria/drug effects , Listeria/genetics , Listeria/classification , Listeria/isolation & purification , Anti-Bacterial Agents/pharmacology , Spain/epidemiology , Listeriosis/microbiology , Listeriosis/veterinary , Listeriosis/epidemiology , Genotype , Drug Resistance, Bacterial/genetics , Whole Genome Sequencing , Listeria monocytogenes/drug effects , Listeria monocytogenes/genetics , Listeria monocytogenes/isolation & purification , Humans , Phenotype
19.
Foodborne Pathog Dis ; 21(7): 424-430, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38597599

ABSTRACT

Listeria monocytogenes is a foodborne pathogen. In 2022, we collected 15 strains of L. monocytogenes isolated from patients in some foodborne disease sentinel monitoring hospitals in Sichuan Province. Through whole genome sequencing (WGS), we obtained the virulence genes carried by the strains, multi-locus sequence typing (MLST), core genome MLST (cgMLST), clonal complex (CC), and serum groups and constructed a phylogenetic tree and minimum spanning tree with nonhuman strains. An analysis shows that all 15 strains of L. monocytogenes carry virulence genes LIPI-1 and LIPI-2, whereas the carrying rates of LIPI-3 and LIPI-4 virulence genes are relatively low. The MLST typing results showed a total of 10 sequence types (ST), including 10 CCs, with ST7 being the dominant type. The cgMLST clearly distinguishes strains of different lineages and CC types. The serum group is divided into three types: IIa, IIb, and IVb, with IIa being the dominant serum group. An analysis of antibiotic genes showed that all 15 strains carried FosX, lin, mprF, and norB with high carrying rates. The minimum inhibitory concentration results indicated that all were susceptible to eight antibiotics (ampicillin, penicillin, tetracycline, meropenem, erythromycin, vancomycin, ciprofloxacin, and trimethoprim-sulfamethoxazole). The analysis of strains isolated from different sources of Listeria revealed varying degrees of diversity, and the contamination of meat and environment within the province is closely related to clinical cases. L. monocytogenes isolated from clinical cases in Sichuan Province carry multiple virulence and antibiotic genes, with high potential pathogenicity. It is necessary to further strengthen the monitoring and control of food and environment by L. monocytogenes within Sichuan Province.


Subject(s)
Anti-Bacterial Agents , Listeria monocytogenes , Listeriosis , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phylogeny , Virulence Factors , Whole Genome Sequencing , Listeria monocytogenes/genetics , Listeria monocytogenes/isolation & purification , Listeria monocytogenes/classification , China , Humans , Anti-Bacterial Agents/pharmacology , Listeriosis/microbiology , Listeriosis/epidemiology , Virulence Factors/genetics , Foodborne Diseases/microbiology , Virulence/genetics , Genome, Bacterial , Food Microbiology
20.
Proc Natl Acad Sci U S A ; 121(17): e2320311121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38635627

ABSTRACT

Listeria monocytogenes is a bacterial pathogen that can cause life-threatening central nervous system (CNS) infections. While mechanisms by which L. monocytogenes and other pathogens traffic to the brain have been studied, a quantitative understanding of the underlying dynamics of colonization and replication within the brain is still lacking. In this study, we used barcoded L. monocytogenes to quantify the bottlenecks and dissemination patterns that lead to cerebral infection. Following intravenous (IV) inoculation, multiple independent invasion events seeded all parts of the CNS from the blood, however, only one clone usually became dominant in the brain. Sequential IV inoculations and intracranial inoculations suggested that clones that had a temporal advantage (i.e., seeded the CNS first), rather than a spatial advantage (i.e., invaded a particular brain region), were the main drivers of clonal dominance. In a foodborne model of cerebral infection with immunocompromised mice, rare invasion events instead led to a highly infected yet monoclonal CNS. This restrictive bottleneck likely arose from pathogen transit into the blood, rather than directly from the blood to the brain. Collectively, our findings provide a detailed quantitative understanding of the L. monocytogenes population dynamics that lead to CNS infection and a framework for studying the dynamics of other cerebral infections.


Subject(s)
Central Nervous System Infections , Listeria monocytogenes , Listeriosis , Mice , Animals , Listeria monocytogenes/physiology , Listeriosis/microbiology , Brain/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL