Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 737
Filter
1.
Placenta ; 128: 29-35, 2022 10.
Article in English | MEDLINE | ID: mdl-36057170

ABSTRACT

INTRODUCTION: Placental infection and inflammation are risk factors for adverse pregnancy outcomes, including preterm labor. However, the mechanisms underlying these outcomes are poorly understood. METHODS: To study this response, we have employed a pregnant mouse model of placental infection caused by the bacterial pathogen Listeria monocyogenes, which infects the human placenta. Through in vivo bioluminescence imaging, we confirm the presence of placental infection and quantify relative infection levels. Infected and control placentas were collected on embryonic day 18 for RNA sequencing to evaluate gene expression signatures associated with infection by Listeria. RESULTS: We identified an enrichment of genes associated with eicosanoid biosynthesis, suggesting an increase in eicosanoid production in infected tissues. Because of the known importance of eicosanoids in inflammation and timing of labor, we quantified eicosanoid levels in infected and uninfected placentas using semi-targeted mass spectrometry. We found a significant increase in the concentrations of several key eicosanoids: leukotriene B4, lipoxin A4, prostaglandin A2, prostaglandin D2, and eicosatrienoic acid. DISCUSSION: Our study provides a likely explanation for dysregulation of the timing of labor following placental infection. Further, our results suggest potential biomarkers of placental pathology and targets for clinical intervention.


Subject(s)
Listeria monocytogenes , Listeriosis , Pregnancy Complications, Infectious , Animals , Biomarkers/metabolism , Female , Humans , Infant, Newborn , Inflammation/metabolism , Leukotriene B4/metabolism , Listeriosis/complications , Listeriosis/microbiology , Listeriosis/pathology , Mice , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/pathology , Prostaglandin D2/metabolism , Transcriptome
2.
Elife ; 112022 04 05.
Article in English | MEDLINE | ID: mdl-35380108

ABSTRACT

Cellular respiration is essential for multiple bacterial pathogens and a validated antibiotic target. In addition to driving oxidative phosphorylation, bacterial respiration has a variety of ancillary functions that obscure its contribution to pathogenesis. We find here that the intracellular pathogen Listeria monocytogenes encodes two respiratory pathways which are partially functionally redundant and indispensable for pathogenesis. Loss of respiration decreased NAD+ regeneration, but this could be specifically reversed by heterologous expression of a water-forming NADH oxidase (NOX). NOX expression fully rescued intracellular growth defects and increased L. monocytogenes loads >1000-fold in a mouse infection model. Consistent with NAD+ regeneration maintaining L. monocytogenes viability and enabling immune evasion, a respiration-deficient strain exhibited elevated bacteriolysis within the host cytosol and NOX expression rescued this phenotype. These studies show that NAD+ regeneration represents a major role of L. monocytogenes respiration and highlight the nuanced relationship between bacterial metabolism, physiology, and pathogenesis.


Cellular respiration is one of the main ways organisms make energy. It works by linking the oxidation of an electron donor (like sugar) to the reduction of an electron acceptor (like oxygen). Electrons pass between the two molecules along what is known as an 'electron transport chain'. This process generates a force that powers the production of adenosine triphosphate (ATP), a molecule that cells use to store energy. Respiration is a common way for cells to replenish their energy stores, but it is not the only way. A simpler process that does not require a separate electron acceptor or an electron transport chain is called fermentation. Many bacteria have the capacity to perform both respiration and fermentation and do so in a context-dependent manner. Research has shown that respiration can contribute to bacterial diseases, like tuberculosis and listeriosis (a disease caused by the foodborne pathogen Listeria monocytogenes). Indeed, some antibiotics even target bacterial respiration. Despite being often discussed in the context of generating ATP, respiration is also important for many other cellular processes, including maintaining the balance of reduced and oxidized nicotinamide adenine dinucleotide (NAD) cofactors. Because of these multiple functions, the exact role respiration plays in disease is unknown. To find out more, Rivera-Lugo, Deng et al. developed strains of the bacterial pathogen Listeria monocytogenes that lacked some of the genes used in respiration. The resulting bacteria were still able to produce energy, but they became much worse at infecting mammalian cells. The use of a genetic tool that restored the balance of reduced and oxidized NAD cofactors revived the ability of respiration-deficient L. monocytogenes to infect mammalian cells, indicating that this balance is what the bacterium requires to infect. Research into respiration tends to focus on its role in generating ATP. But these results show that for some bacteria, this might not be the most important part of the process. Understanding the other roles of respiration could change the way that researchers develop antibacterial drugs in the future. This in turn could help with the growing problem of antibiotic resistance.


Subject(s)
Listeria monocytogenes , Listeriosis , Animals , Cell Respiration , Disease Models, Animal , Immune Evasion , Listeria monocytogenes/genetics , Listeriosis/metabolism , Listeriosis/microbiology , Listeriosis/pathology , Mice , NAD/metabolism
3.
PLoS Pathog ; 17(8): e1009379, 2021 08.
Article in English | MEDLINE | ID: mdl-34398937

ABSTRACT

The Gram-positive bacterium Listeria monocytogenes is the causative agent of the foodborne disease listeriosis, one of the deadliest bacterial infections known. In order to cause disease, L. monocytogenes must properly coordinate its metabolic and virulence programs in response to rapidly changing environments within the host. However, the mechanisms by which L. monocytogenes senses and adapts to the many stressors encountered as it transits through the gastrointestinal (GI) tract and disseminates to peripheral organs are not well understood. In this study, we investigated the role of the redox-responsive transcriptional regulator Rex in L. monocytogenes growth and pathogenesis. Rex is a conserved canonical transcriptional repressor that monitors the intracellular redox state of the cell by sensing the ratio of reduced and oxidized nicotinamide adenine dinucleotides (NADH and NAD+, respectively). Here, we demonstrated that L. monocytogenes Rex represses fermentative metabolism and is therefore required for optimal growth in the presence of oxygen. We also show that in vitro, Rex represses the production of virulence factors required for survival and invasion of the GI tract, as a strain lacking rex was more resistant to acidified bile and invaded host cells better than wild type. Consistent with these results, Rex was dispensable for colonizing the GI tract and disseminating to peripheral organs in an oral listeriosis model of infection. However, Rex-dependent regulation was required for colonizing the spleen and liver, and L. monocytogenes lacking the Rex repressor were nearly sterilized from the gallbladder. Taken together, these results demonstrated that Rex functions as a repressor of fermentative metabolism and suggests a role for Rex-dependent regulation in L. monocytogenes pathogenesis. Importantly, the gallbladder is the bacterial reservoir during listeriosis, and our data suggest redox sensing and Rex-dependent regulation are necessary for bacterial survival and replication in this organ.


Subject(s)
Bacterial Proteins/metabolism , Fermentation , Gene Products, rex/metabolism , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Virulence Factors/metabolism , Virulence , Animals , Bacterial Proteins/genetics , Female , Gene Expression Regulation, Bacterial , Gene Products, rex/genetics , Listeriosis/metabolism , Listeriosis/pathology , Mice , Mice, Inbred BALB C , Oxidation-Reduction , Transcriptome , Virulence Factors/genetics
4.
Nat Immunol ; 22(6): 699-710, 2021 06.
Article in English | MEDLINE | ID: mdl-34040226

ABSTRACT

It is increasingly recognized that immune development within mucosal tissues is under the control of environmental factors during early life. However, the cellular mechanisms that underlie such temporally and regionally restrictive governance of these processes are unclear. Here, we uncover an extrathymic pathway of immune development within the colon that is controlled by embryonic but not bone marrow-derived macrophages, which determines the ability of these organs to receive invariant natural killer T (iNKT) cells and allow them to establish local residency. Consequently, early-life perturbations of fetal-derived macrophages result in persistent decreases of mucosal iNKT cells and is associated with later-life susceptibility or resistance to iNKT cell-associated mucosal disorders. These studies uncover a host developmental program orchestrated by ontogenically distinct macrophages that is regulated by microbiota, and they reveal an important postnatal function of macrophages that emerge in fetal life.


Subject(s)
Colitis/immunology , Intestinal Mucosa/immunology , Listeriosis/immunology , Macrophages/immunology , Mucosal-Associated Invariant T Cells/immunology , Animals , Cell Proliferation/genetics , Colitis/microbiology , Colitis/pathology , Colon/cytology , Colon/embryology , Colon/immunology , Colon/pathology , Cytokines/metabolism , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Embryo, Mammalian , Female , Gastrointestinal Microbiome/immunology , Gene Expression Regulation, Developmental/immunology , Germ-Free Life , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestinal Mucosa/pathology , Listeriosis/microbiology , Listeriosis/pathology , Macrophages/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , RNA-Seq , Signal Transduction/genetics , Signal Transduction/immunology
5.
PLoS Pathog ; 17(4): e1009531, 2021 04.
Article in English | MEDLINE | ID: mdl-33878120

ABSTRACT

Most individuals who consume foods contaminated with the bacterial pathogen Listeria monocytogenes (Lm) develop mild symptoms, while others are susceptible to life-threatening systemic infections (listeriosis). Although it is known that the risk of severe disease is increased in certain human populations, including the elderly, it remains unclear why others who consume contaminated food develop listeriosis. Here, we used a murine model to discover that pulmonary coinfections can impair the host's ability to adequately control and eradicate systemic Lm that cross from the intestines to the bloodstream. We found that the resistance of mice to oral Lm infection was dramatically reduced by coinfection with Streptococcus pneumoniae (Spn), a bacterium that colonizes the respiratory tract and can also cause severe infections in the elderly. Exposure to Spn or microbial products, including a recombinant Lm protein (L1S) and lipopolysaccharide (LPS), rendered otherwise resistant hosts susceptible to severe systemic Lm infection. In addition, we show that this increase in susceptibility was dependent on an increase in the production of interleukin-10 (IL-10) from Ncr1+ cells, including natural killer (NK) cells. Lastly, the ability of Ncr1+ cell derived IL-10 to increase disease susceptibility correlated with a dampening of both myeloid cell accumulation and myeloid cell phagocytic capacity in infected tissues. These data suggest that efforts to minimize inflammation in response to an insult at the respiratory mucosa render the host more susceptible to infections by Lm and possibly other pathogens that access the oral mucosa.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , Pneumonia/immunology , Animals , Disease Progression , Disease Susceptibility , Female , Interleukin-10/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Lipopolysaccharides , Listeria monocytogenes/pathogenicity , Listeriosis/complications , Listeriosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouth Diseases/complications , Mouth Diseases/immunology , Mouth Diseases/microbiology , Mouth Diseases/pathology , Pneumonia/complications , Pneumonia/etiology , Pneumonia/pathology
6.
Front Cell Infect Microbiol ; 11: 748461, 2021.
Article in English | MEDLINE | ID: mdl-35223532

ABSTRACT

The internalin family proteins, which carry the leucine repeat region structural motif, play diverse roles in Listeria monocytogenes (Lm) infection and pathogenesis. Although Internalin F, encoded by inlF, was identified more than 20 years ago, its role in the Lm anti-inflammatory response remains unknown. Lm serotype 4b isolates are associated with the majority of listeriosis outbreaks, but the function of InlF in these strains is not fully understood. In this study, we aimed to elucidate the role of inlF in modulating the inflammatory response and pathogenesis of the 4b strain Lm NTSN. Strikingly, although inlF was highly expressed at the transcriptional level during infection of five non-phagocytic cell types, it was not involved in adherence or invasion. Conversely, inlF did contributed to Lm adhesion and invasion of macrophages, and dramatically suppressed the expression of pro-inflammatory cytokines interleukin (IL)-1ß and tumor necrosis factor (TNF-α). Consistent with the in vitro results, during Lm infection mice, inlF significantly inhibited the expression of IL-1ß and IL-6 in the spleen, as well as IL-1ß, IL-6, and TNF-α in the liver. More importantly, inlF contributed to Lm colonization in the spleen, liver, and ileum during the early stage of mouse infection via intragastric administration, inducing severe inflammatory injury and histopathologic changes in the late stage. To our knowledge, this is the first report to demonstrate that inlF mediates the inhibition of the pro-inflammatory response and contributes to the colonization and survival of Lm during the early stage of infection in mice. Our research partly explains the high pathogenicity of serovar 4b strains and will lead to new insights into the pathogenesis and immune evasion of Lm.


Subject(s)
Bacterial Proteins , Listeria monocytogenes , Listeriosis , Animals , Cytokines , Listeriosis/pathology , Mice , Serogroup , Virulence
7.
Methods Mol Biol ; 2220: 189-200, 2021.
Article in English | MEDLINE | ID: mdl-32975776

ABSTRACT

Listeria monocytogenes is a model intracellular pathogen that can invade the cytoplasm of host mammalian cells. Cellular invasion can be measured using standard techniques, such as the classical gentamicin protection assay, based on the quantification of colony-forming units from lysates of infected cells. In addition, there are methods based on immunofluorescence microscopy which allow for assaying invasion in a medium- to high-throughput manner. In the following sections, we detail two different assays that can be used alone or in combination to quantify the internalization of L. monocytogenes in host cells.


Subject(s)
Listeria monocytogenes/physiology , Listeriosis/pathology , Bacterial Load/methods , Colony Count, Microbial/methods , HeLa Cells , Host-Pathogen Interactions , Humans , Listeriosis/microbiology , Staining and Labeling/methods
8.
Methods Mol Biol ; 2220: 201-215, 2021.
Article in English | MEDLINE | ID: mdl-32975777

ABSTRACT

The pathogen Listeria monocytogenes is a facultative intracellular bacterium, which targets a large range of cell types. Following entry, bacteria disrupt the invasion vacuole and reach the cytoplasm where they replicate and use the actin cytoskeleton to propel themselves from cell to cell. Mammalian epithelial cells grown in vitro can be used to study the different steps of the intracellular life of Listeria. However, rapid multiplication and dissemination of bacteria can induce important cell death and detachment, resulting in the formation of lytic plaques. Thus, in vitro infections with L. monocytogenes are usually restricted to short time courses, from a few minutes to one day. Here, we present a method to study long-term L. monocytogenes infections in epithelial cells using epifluorescence microscopy. This protocol enables the observation of actin-based motility, intercellular dissemination foci, and entrapment of L. monocytogenes within vacuoles of persistence termed "Listeria-Containing Vacuoles" (LisCVs). We also describe a protocol to study the recruitment of cytoskeletal proteins at Listeria actin comet tails, as well as a method to assess the membrane integrity of intracellular bacteria using a LIVE/DEAD viability assay.


Subject(s)
Epithelial Cells/microbiology , Listeria monocytogenes/physiology , Listeriosis/pathology , Microscopy, Fluorescence/methods , Cell Line , Cytoskeletal Proteins/analysis , Epithelial Cells/pathology , Fluorescent Antibody Technique/methods , Host-Pathogen Interactions , Humans , Listeria monocytogenes/isolation & purification , Listeriosis/microbiology
9.
J Exp Med ; 218(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-32976558

ABSTRACT

The human placenta is a dynamic organ that modulates physiological adaptations to pregnancy. To define the immunological signature of the human placenta, we performed unbiased profiling of secreted immune factors from human chorionic villi isolated from placentas at mid and late stages of pregnancy. We show that placental trophoblasts constitutively secrete the inflammasome-associated cytokines IL-1ß and IL-18, which is blocked by NLRP3 inflammasome inhibitors and occurs without detectable gasdermin D cleavage. We further show that placenta-derived IL-1ß primes monocytes for inflammasome induction to protect against Listeria monocytogenes infection. Last, we show that the human placenta responds to L. monocytogenes infection through additional inflammasome activation and that inhibition of this pathway sensitizes villi to infection. Our results thus identify the inflammasome as an important mechanism by which the human placenta regulates systemic and local immunity during pregnancy to defend against L. monocytogenes infection.


Subject(s)
Chorionic Villi/immunology , Inflammasomes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Signal Transduction/immunology , Trophoblasts/immunology , Caco-2 Cells , Chorionic Villi/microbiology , Chorionic Villi/pathology , Female , Humans , Interleukin-18/immunology , Interleukin-1beta/immunology , Listeriosis/microbiology , Listeriosis/pathology , Monocytes/immunology , Monocytes/microbiology , Monocytes/pathology , THP-1 Cells , Trophoblasts/microbiology , Trophoblasts/pathology
10.
Front Immunol ; 11: 597433, 2020.
Article in English | MEDLINE | ID: mdl-33329589

ABSTRACT

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Subject(s)
Antigens, CD/immunology , Erythroid Precursor Cells , Immunosuppression Therapy , Listeria monocytogenes/immunology , Listeriosis , Receptors, Transferrin/immunology , Animals , Animals, Newborn , COVID-19/immunology , COVID-19/pathology , Erythroid Precursor Cells/immunology , Erythroid Precursor Cells/pathology , Erythroid Precursor Cells/transplantation , Female , Heterografts , Humans , Infant, Newborn , Listeriosis/immunology , Listeriosis/pathology , Listeriosis/therapy , Male , Mice , Mice, Inbred BALB C , SARS-CoV-2/immunology
11.
Curr Protoc Immunol ; 130(1): e102, 2020 09.
Article in English | MEDLINE | ID: mdl-32710703

ABSTRACT

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.


Subject(s)
Listeria monocytogenes/physiology , Listeriosis/etiology , Listeriosis/pathology , Liver/microbiology , Liver/pathology , Monocytes/pathology , Neutrophils/pathology , Animals , Biomarkers , Biopsy , Disease Models, Animal , Disease Susceptibility , Female , Foodborne Diseases/microbiology , Humans , Immunophenotyping , Listeriosis/metabolism , Listeriosis/transmission , Mice , Monocytes/immunology , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Species Specificity
12.
Toxins (Basel) ; 12(6)2020 06 20.
Article in English | MEDLINE | ID: mdl-32575670

ABSTRACT

Listeria monocytogenes is among the best-characterized intracellular pathogens. Its virulence factors, and the way they interfere with host cells to hijack host functions and promote the establishment and dissemination of the infection, have been the focus of multiple studies over the last 30 years. During cellular infection, L. monocytogenes was shown to induce host DNA damage and delay the host cell cycle to its own benefit. However, whether the cell cycle stage would interfere with the capacity of Listeria to infect human cultured cell lines was never assessed. We found here that L. monocytogenes preferentially infects cultured cells in G2/M phases. Inside G2/M cells, the bacteria lead to an increase in the overall mitosis duration by delaying the mitotic exit. We showed that L. monocytogenes infection causes a sustained activation of the spindle assembly checkpoint, which we correlated with the increase in the percentage of misaligned chromosomes detected in infected cells. Moreover, we demonstrated that chromosome misalignment in Listeria-infected cells required the function of two Listeria virulence factors, ActA and InlC. Our findings show the pleiotropic role of Listeria virulence factors and their cooperative action in successfully establishing the cellular infection.


Subject(s)
Bacterial Proteins/metabolism , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Membrane Proteins/metabolism , Mitosis , Virulence Factors/metabolism , Bacterial Proteins/genetics , Caco-2 Cells , Chromosome Segregation , G2 Phase Cell Cycle Checkpoints , Host-Pathogen Interactions , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/pathogenicity , Listeriosis/pathology , M Phase Cell Cycle Checkpoints , Membrane Proteins/genetics , Virulence , Virulence Factors/genetics
13.
J Immunol ; 205(3): 760-766, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32540996

ABSTRACT

P2X5 is a member of the P2X purinergic receptor family of ligand-gated cation channels and has recently been shown to regulate inflammatory bone loss. In this study, we report that P2X5 is a protective immune regulator during Listeria monocytogenes infection, as P2X5-deficient mice exhibit increased bacterial loads in the spleen and liver, increased tissue damage, and early (within 3-6 d) susceptibility to systemic L. monocytogenes infection. Whereas P2X5-deficient mice experience normal monocyte recruitment in response to L. monocytogenes, P2X5-deficient bone marrow-derived macrophages (BMMs) exhibit defective cytosolic killing of L. monocytogenes We further showed that P2X5 is required for L. monocytogenes-induced inflammasome activation and IL-1ß production and that defective L. monocytogenes killing in P2X5-deficient BMMs is substantially rescued by exogenous IL-1ß or IL-18. Finally, in vitro BMM killing and in vivo L. monocytogenes infection experiments employing either P2X7 deficiency or extracellular ATP depletion suggest that P2X5-dependent anti-L. monocytogenes immunity is independent of the ATP-P2X7 inflammasome activation pathway. Together, our findings elucidate a novel and specific role for P2X5 as a critical mediator of protective immunity.


Subject(s)
Inflammasomes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/immunology , Monocytes/immunology , Receptors, Purinergic P2X5/deficiency , Adenosine Triphosphate/genetics , Adenosine Triphosphate/immunology , Animals , Disease Susceptibility , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Listeriosis/genetics , Listeriosis/pathology , Macrophages/pathology , Mice , Mice, Knockout , Monocytes/pathology , Receptors, Purinergic P2X5/immunology
14.
J Immunol ; 205(3): 853-863, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591396

ABSTRACT

As a specialized subset of intestinal epithelial cells (IECs), goblet cells (GCs) play an important role during the antibacterial response via mucin production. However, the regulatory mechanisms involved in GC differentiation and function during infection, particularly the role of immune cell-IEC cross-talk, remain largely unknown. In this study, using Villin∆Ltbr conditional knockout mice, we demonstrate that LTßR, expressed on IECs, is required for GC hyperplasia and mucin 2 (MUC2) expression during Listeria infection for host defense but not homeostatic maintenance in the naive state. Analysis of single gene-deficient mice revealed that the ligand lymphotoxin (LT), but not LIGHT, and type 3 innate lymphoid cells (ILC3s), but not conventional T cells, are required for MUC2-dependent Listeria control. Conditional deficiency of LT in ILC3s further confirmed the importance of LT signals derived from ILC3s. Lack of ILC3-derived LT or IEC-derived LTßR resulted in the defective expression of genes related to GC differentiation but was not correlated with IEC proliferation and cell death, which were found to be normal by Ki-67 and Annexin V staining. In addition, the alternative NF-κB signaling pathway (involving RelB) in IECs was found to be required for the expression of GC differentiation-related genes and Muc2 and required for the anti-Listeria response. Therefore, our data together suggest a previously unrecognized ILC3-IEC interaction and LT-LTßR-RelB signaling axis governing GC differentiation and function during Listeria infection for host defense.


Subject(s)
Cell Differentiation/immunology , Goblet Cells/immunology , Listeria/immunology , Listeriosis/immunology , Lymphocytes/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Goblet Cells/pathology , Listeriosis/genetics , Listeriosis/pathology , Lymphocytes/pathology , Lymphotoxin beta Receptor , Lymphotoxin-alpha/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/genetics
15.
PLoS One ; 15(4): e0231393, 2020.
Article in English | MEDLINE | ID: mdl-32352974

ABSTRACT

Whole genome sequencing (WGS) was performed on 201 Listeria monocytogenes isolates recovered from 102 of 27,389 refrigerated ready-to-eat (RTE) food samples purchased at retail in U.S. FoodNet sites as part of the 2010-2013 interagency L. monocytogenes Market Basket Survey (Lm MBS). Core genome multi-locus sequence typing (cgMLST) and in-silico analyses were conducted, and these data were analyzed with metadata for isolates from five food groups: produce, seafood, dairy, meat, and combination foods. Six of 201 isolates, from 3 samples, were subsequently confirmed as L. welshimeri. Three samples contained one isolate per sample; mmong the 96 samples that contained two isolates per sample, 3 samples each contained two different strains and 93 samples each contained duplicate isolates. After 93 duplicate isolates were removed, the remaining 102 isolates were delineated into 29 clonal complexes (CCs) or singletons based on their sequence type. The five most prevalent CCs were CC155, CC1, CC5, CC87, and CC321. The Shannon's diversity index for clones per food group ranged from 1.49 for dairy to 2.32 for produce isolates, which were not significantly different in pairwise comparisons. The most common molecular serogroup as determined by in-silico analysis was IIa (45.6%), followed by IIb (27.2%), IVb (20.4%), and IIc (4.9%). The proportions of isolates within lineages I, II, and III were 48.0%, 50.0% and 2.0%, respectively. Full-length inlA was present in 89.3% of isolates. Listeria pathogenicity island 3 (LIPI-3) and LIPI-4 were found in 51% and 30.6% of lineage I isolates, respectively. Stress survival islet 1 (SSI-1) was present in 34.7% of lineage I isolates, 80.4% of lineage II isolates and the 2 lineage III isolates; SSI-2 was present only in the CC121 isolate. Plasmids were found in 48% of isolates, including 24.5% of lineage I isolates and 72.5% of lineage II isolates. Among the plasmid-carrying isolates, 100% contained at least one cadmium resistance cassette and 89.8% contained bcrABC, involved in quaternary ammonium compound tolerance. Multiple clusters of isolates from different food samples were identified by cgMLST which, along with available metadata, could aid in the investigation of possible cross-contamination and persistence events.


Subject(s)
Food Microbiology , Genetic Variation , Listeria monocytogenes/genetics , Virulence/genetics , Bacterial Proteins/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/metabolism , Humans , Listeria monocytogenes/classification , Listeria monocytogenes/isolation & purification , Listeria monocytogenes/pathogenicity , Listeriosis/pathology , Listeriosis/transmission , Multilocus Sequence Typing , Phylogeny , Plasmids/genetics , Plasmids/metabolism , Serogroup , Whole Genome Sequencing
16.
PLoS Pathog ; 16(5): e1008497, 2020 05.
Article in English | MEDLINE | ID: mdl-32453780

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) are at the forefront of host-microbe interactions. Molecular and cell-based studies suggest that HSPG-pathogen interactions promote pathogenesis by facilitating microbial attachment and invasion of host cells. However, the specific identity of HSPGs, precise mechanisms by which HSPGs promote pathogenesis, and the in vivo relevance of HSPG-pathogen interactions remain to be determined. HSPGs also modulate host responses to tissue injury and inflammation, but functions of HSPGs other than facilitating microbial attachment and internalization are understudied in infectious disease. Here we examined the role of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, in mouse models of Listeria monocytogenes (Lm) infection. We show that Sdc1-/- mice are significantly less susceptible to both intragastric and intravenous Lm infection compared to wild type (Wt) mice. This phenotype is not seen in Sdc3-/- or Sdc4-/- mice, indicating that ablation of Sdc1 causes a specific gain of function that enables mice to resist listeriosis. However, Sdc1 does not support Lm attachment or invasion of host cells, indicating that Sdc1 does not promote pathogenesis as a cell surface Lm receptor. Instead, Sdc1 inhibits the clearance of Lm before the bacterium gains access to its intracellular niche. Large intravascular aggregates of neutrophils and neutrophil extracellular traps (NETs) embedded with antimicrobial compounds are formed in Sdc1-/- livers, which trap and kill Lm. Lm infection induces Sdc1 shedding from the surface of hepatocytes in Wt livers, which is directly associated with the decrease in size of intravascular aggregated NETs. Furthermore, administration of purified Sdc1 ectodomains or DNase inhibits the formation of intravascular aggregated neutrophils and NETs and significantly increases the liver bacterial burden in Sdc1-/- mice. These data indicate that Lm induces Sdc1 shedding to subvert the activity of Sdc1 ectodomains to inhibit its clearance by intravascular aggregated NETs.


Subject(s)
Extracellular Traps/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Neutrophils/immunology , Syndecan-1/immunology , Animals , Extracellular Traps/genetics , Hepatocytes/immunology , Hepatocytes/pathology , Listeria monocytogenes/pathogenicity , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/pathology , Protein Domains , Syndecan-1/genetics
18.
Toxins (Basel) ; 12(5)2020 05 05.
Article in English | MEDLINE | ID: mdl-32380697

ABSTRACT

Although it rarely induces disease in humans, Listeria monocytogenes (Lm) is important due to the frequency of serious pathological conditions-such as sepsis and meningitis-it causes in those few people that do get infected. Virulence factors (VF) of Lm-especially those involved in the passage through multiple cellular barriers of the body, including internalin (Inl) family members and listeriolysin O (LLO)-have been investigated both in vitro and in vivo, but the majority of work was focused on the mechanisms utilized during penetration of the gut and fetoplacental barriers. The role of listerial VF during entry into other organs remain as only partially solved puzzles. Here, we review the current knowledge on the entry of Lm into one of its more significant destinations, the brain, with a specific focus on the role of various VF in cellular adhesion and invasion.


Subject(s)
Brain/microbiology , Central Nervous System Bacterial Infections/microbiology , Listeria monocytogenes/metabolism , Listeriosis/microbiology , Virulence Factors/metabolism , Animals , Bacterial Adhesion , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Brain/metabolism , Brain/pathology , Central Nervous System Bacterial Infections/metabolism , Central Nervous System Bacterial Infections/pathology , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Host-Pathogen Interactions , Humans , Listeria monocytogenes/pathogenicity , Listeriosis/metabolism , Listeriosis/pathology , Virulence
20.
Int J Biol Macromol ; 150: 802-813, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32057883

ABSTRACT

Infectious diseases such as Listeria monocytogenes infection pose a great threat to the health of human beings and the development of livestock and poultry farming. Currently the treatment of Listeria infection mainly relies on antibiotics, which may result in excessive antibiotic residues in livestock and poultry products, as well as causing an increase in the occurrence of zoonotic diseases. Here, we demonstrate that Succinoglycan Riclin promoted the clearance of Listeria in the in vitro and in vivo infection model. The expression and secretion of inflammatory cytokines including IL-6 and IL-1ß were significantly increased after Riclin treatment upon infection. The protective effect of Riclin was mainly through activating MAPK/IL-6 axis. HO-1/IL-1ß signaling pathway was less important in this process. Moreover, Riclin caused significant metabolic changes including pathways involved in glycolysis, protein synthesis and oxidative stress during Listeria infection. These results suggest a potential use of Succinoglycan Riclin as non-antibiotic preventive and therapeutic anti-microbial agent in livestock and poultry farming and human diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Interleukin-6/metabolism , Listeria monocytogenes/metabolism , Listeriosis/drug therapy , MAP Kinase Signaling System/drug effects , Polysaccharides, Bacterial/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Listeriosis/metabolism , Listeriosis/pathology , Mice , Polysaccharides, Bacterial/chemistry , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...