Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 211
Filter
Add more filters

Publication year range
1.
PLoS Pathog ; 17(10): e1009987, 2021 10.
Article in English | MEDLINE | ID: mdl-34669717

ABSTRACT

Salmonella enterica represents over 2500 serovars associated with a wide-ranging spectrum of disease; from self-limiting gastroenteritis to invasive infections caused by non-typhoidal serovars (NTS) and typhoidal serovars, respectively. Host factors strongly influence infection outcome as malnourished or immunocompromised individuals can develop invasive infections from NTS, however, comparative analyses of serovar-specific host responses have been constrained by reliance on limited model systems. Here we used human intestinal organoids (HIOs), a three-dimensional "gut-like" in vitro system derived from human embryonic stem cells, to elucidate similarities and differences in host responses to NTS and typhoidal serovars. HIOs discriminated between the two most prevalent NTS, Salmonella enterica serovar Typhimurium (STM) and Salmonella enterica serovar Enteritidis (SE), and typhoidal serovar Salmonella enterica serovar Typhi (ST) in epithelial cell invasion, replication and transcriptional responses. Pro-inflammatory signaling and cytokine output was reduced in ST-infected HIOs compared to NTS infections, consistent with early stages of NTS and typhoidal diseases. While we predicted that ST would induce a distinct transcriptional profile from the NTS strains, more nuanced expression profiles emerged. Notably, pathways involved in cell cycle, metabolism and mitochondrial functions were downregulated in STM-infected HIOs and upregulated in SE-infected HIOs. These results correlated with suppression of cellular proliferation and induction of host cell death in STM-infected HIOs and in contrast, elevated levels of reactive oxygen species production in SE-infected HIOs. Collectively, these results suggest that the HIO model is well suited to reveal host transcriptional programming specific to infection by individual Salmonella serovars, and that individual NTS may provoke unique host epithelial responses during intestinal stages of infection.


Subject(s)
Gene Expression Profiling , Intestines/microbiology , Intestines/physiopathology , Salmonella Infections/microbiology , Salmonella Infections/physiopathology , Humans , Organoids , Salmonella enterica , Serogroup , Transcriptome
2.
PLoS Biol ; 17(4): e3000044, 2019 04.
Article in English | MEDLINE | ID: mdl-30964858

ABSTRACT

Many bacterial pathogens hijack macrophages to egress from the port of entry to the lymphatic drainage and/or bloodstream, causing dissemination of life-threatening infections. However, the underlying mechanisms are not well understood. Here, we report that Salmonella infection generates directional electric fields (EFs) in the follicle-associated epithelium of mouse cecum. In vitro application of an EF, mimicking the infection-generated electric field (IGEF), induces directional migration of primary mouse macrophages to the anode, which is reversed to the cathode upon Salmonella infection. This infection-dependent directional switch is independent of the Salmonella pathogenicity island 1 (SPI-1) type III secretion system. The switch is accompanied by a reduction of sialic acids on glycosylated surface components during phagocytosis of bacteria, which is absent in macrophages challenged by microspheres. Moreover, enzymatic cleavage of terminally exposed sialic acids reduces macrophage surface negativity and severely impairs directional migration of macrophages in response to an EF. Based on these findings, we propose that macrophages are attracted to the site of infection by a combination of chemotaxis and galvanotaxis; after phagocytosis of bacteria, surface electrical properties of the macrophage change, and galvanotaxis directs the cells away from the site of infection.


Subject(s)
Gastrointestinal Tract/immunology , Macrophages/physiology , Taxis Response/physiology , Animals , Bacterial Proteins , Cell Movement/physiology , Electric Conductivity , Electricity , Epithelium/immunology , Epithelium/metabolism , Female , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Phagocytosis , Salmonella/pathogenicity , Salmonella Infections/metabolism , Salmonella Infections/physiopathology
3.
BMC Cardiovasc Disord ; 20(1): 406, 2020 09 07.
Article in English | MEDLINE | ID: mdl-32894058

ABSTRACT

BACKGROUND: Infectious aortic aneurysm, defined as a focal dilation of an infectious arterial wall, is an uncommon life-threatening disease. Compared with open surgery, endovascular repair yields acceptable clinical outcomes. However, residual tissue infection may increase the risk of secondary intervention. Here, we present a successful case of endovascular repair combined with staged drainage for the treatment of infectious aortic aneurysm. CASE PRESENTATION: A 58-year-old man presented to hospital with a 3-day history of lower back pain radiating to the back associated with fever. The dynamic imaging characteristics revealed rapid progress of infectious abdominal aortic aneurysm with negative blood culture. The patient underwent endovascular repair and salmonella enteritidis was identified through drain culture. CONCLUSIONS: Endovascular procedure and staged drainage can be feasible and effective option in selected cases.


Subject(s)
Aneurysm, Infected/surgery , Aortic Aneurysm, Abdominal/surgery , Blood Vessel Prosthesis Implantation , Drainage , Endovascular Procedures , Salmonella Infections/surgery , Salmonella enteritidis/isolation & purification , Aneurysm, Infected/diagnostic imaging , Aneurysm, Infected/microbiology , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/microbiology , Humans , Male , Middle Aged , Salmonella Infections/diagnostic imaging , Salmonella Infections/physiopathology , Treatment Outcome
4.
J Transl Med ; 16(1): 239, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30165857

ABSTRACT

Non-typhoidal Salmonella usually induces self-limiting gastroenteritis. However, in many parts of Africa, especially in individuals who are malnourished, infected with malaria, or have sickle cell disease, the organism causes serious and potentially fatal systemic infections. Since the portal of entry of non-typhoidal Salmonella into the systemic circulation is by way of the intestine, we argue that an increased gut permeability plays a vital role in the initiation of invasive non-typhoidal Salmonella in these patients. Here, we will appraise the evidence supporting a breach in the intestinal barrier and propose the mechanisms for the increased risks for invasive non-typhoidal Salmonella infections in these individuals.


Subject(s)
Anemia, Sickle Cell/complications , Gastrointestinal Microbiome , Intestines/pathology , Salmonella Infections/complications , Salmonella Infections/physiopathology , Africa , Anemia, Sickle Cell/microbiology , Anti-Bacterial Agents/therapeutic use , Humans , Malaria/complications , Malnutrition/complications , Models, Theoretical , Permeability , Risk , Salmonella , Typhoid Fever
5.
J Therm Biol ; 73: 8-13, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29549994

ABSTRACT

Behavioral fever in reptiles is often considered an adaptive response used to eliminate pathogens, yet empirical data showing the wide-spread use of this response is mixed. This behavioral change can be beneficial by enhancing the host's immune response and increasing the animal's chance of survival, but it can also be detrimental in terms of host energetic requirements and enzymatic performance. Thus, we examined whether captive-bred African house snakes (Lamprophis fuliginosus) employed behavioral fever in response to pathogen stimulus. Twenty-one African house snakes were injected separately with three different strains of ultraviolet (UV) light-killed bacteria (Escherichia coli, Staphylococcus aureus, Salmonella enterica). We found an increased variance of hourly cloacal temperatures following exposure to pathogens in male but not female house snakes. We did not, however, find a significant febrile response to pathogen exposure as measured via mean cloacal temperature. This research adds critical information to the field of reptilian physiology as this field remains understudied. Reptilian immune function and its relationship with thermal biology is ever more pertinent as new challenges arise, such as novel pathogens and changing climate.


Subject(s)
Bacterial Infections/physiopathology , Body Temperature Regulation , Sex Characteristics , Snakes/physiology , Animals , Cloaca/physiopathology , Escherichia coli/pathogenicity , Escherichia coli Infections/physiopathology , Female , Male , Salmonella Infections/physiopathology , Salmonella enterica/pathogenicity , Staphylococcal Infections/physiopathology , Staphylococcus aureus/pathogenicity
6.
J Physiol ; 595(2): 423-432, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27284010

ABSTRACT

Transport of fluid and electrolytes in the intestine allows for appropriate adjustments in luminal fluidity while reclaiming water used in digesting and absorbing a meal, and is closely regulated. This article discusses various endogenous and exogenous mechanisms whereby transport is controlled in the gut, placing these in the context of the ideas about the neurohumoral control of alimentary physiology that were promulgated by William Bayliss and Ernest Starling. The article considers three themes. First, mechanisms that intrinsically regulate chloride secretion, centred on the epidermal growth factor receptor (EGFr), are discussed. These may be important in ensuring that excessive chloride secretion, with the accompanying loss of fluid, is not normally stimulated by intestinal distension as the meal passes through the gastrointestinal tract. Second, mechanisms whereby probiotic microorganisms can impart beneficial effects on the gut are described, with a focus on targets at the level of the epithelium. These findings imply that the commensal microbiota exert important influences on the epithelium in health and disease. Finally, mechanisms that lead to diarrhoea in patients infected with an invasive pathogen, Salmonella, are considered, based on recent studies in a novel mouse model. Diarrhoea is most likely attributable to reduced expression of absorptive transporters and may not require the influx of neutrophils that accompanies infection. Overall, the goal of the article is to highlight the many ways in which critical functions of the intestinal epithelium are regulated under physiological and pathophysiological conditions, and to suggest possible targets for new therapies for digestive disease states.


Subject(s)
Epithelial Cells/physiology , Gastrointestinal Microbiome , Gastrointestinal Tract/physiology , Animals , Chlorides/physiology , Diarrhea/physiopathology , Gastrointestinal Tract/microbiology , Humans , Probiotics , Salmonella Infections/physiopathology
7.
Cytokine ; 98: 27-32, 2017 10.
Article in English | MEDLINE | ID: mdl-27773552

ABSTRACT

Salmonella enterica is a facultative intracellular bacterium that is the leading cause of food borne illnesses in humans. The cytokine IFN-γ has well-established antibacterial properties against Salmonella and other intracellular microbes, for example its capacity to activate macrophages, promote phagocytosis, and destroy phagocytosed microbes by free radical-driven toxification of phagosomes. But IFN-γ induces the expression of hundreds of uncharacterized genes, suggesting that this cytokine deploys additional antimicrobial strategies that await discovery. Recently, one such mechanism, mediated by a family of IFN-inducible small GTPases called Guanylate Binding Proteins (GBPs) has been uncovered. GBPs were shown to facilitate the pyroptotic clearance of Salmonella from infected macrophages by rupturing the protective intracellular vacuole this microbe forms around itself. Once this protective vacuole is lost, exposed Salmonella activates pyroptosis, which destroys the infected cell. In this review, we summarize such emerging roles for IFN-γ in restricting Salmonella pathogenesis.


Subject(s)
Interferon-gamma/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Animals , Autophagy , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Inflammasomes , Macrophages/immunology , Macrophages/microbiology , Mice , Phagocytosis , Phagosomes/immunology , Phagosomes/microbiology , Pyroptosis , Salmonella Infections/microbiology , Salmonella Infections/physiopathology
8.
J Nutr ; 146(8): 1492-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27358412

ABSTRACT

BACKGROUND: One promising strategy for reducing human salmonellosis induced by Salmonella Enteritidis is to supplement animal diets with natural feed additives such as mannan oligosaccharides (MOSs). OBJECTIVE: We sought to investigate the potential role of Salmosan (S-ßGM), an MOS product extremely rich in ß-galactomannan, in preventing epithelial barrier function disruption induced by S. Enteritidis colonization in an in vitro model of intestinal Caco-2 cells in culture. METHODS: Differentiated Caco-2 cells were incubated for 3 h with S. Enteritidis at a multiplicity of infection of 10 in the absence or presence of 500 µg S-ßGM/mL. Paracellular permeability (PP) was assessed by transepithelial electrical resistance (TER), d-mannitol, and fluorescein isothiocyanate-dextran (FD-4) flux. Tight junction proteins and cytoskeletal actin were also localized by confocal microscopy. Reactive oxygen species (ROS) and lipid peroxidation products were evaluated. Scanning and transmission electron microscopy were used to visualize S. Enteritidis adhesion to, and invasion of, the Caco-2 cell cultures. RESULTS: Compared with controls, TER was significantly reduced by 30%, and d-mannitol and FD-4 flux were significantly increased by 374% and 54% in S. Enteritidis-infected cultures, respectively. The presence of S-ßGM in infected cultures induced total recoveries of TER and FD-4 flux to values that did not differ from the control and a partial recovery of d-mannitol flux. These effects were confirmed by immunolocalization of actin, zonula occludens protein 1, and occludin. Similar results were obtained for Salmonella Dublin. The protection of S-ßGM on PP in infected cultures may be associated with a total recovery of ROS production to values that did not differ from the control. Moreover, S-ßGM has the capacity to agglutinate bacteria, leading to a significant reduction of 32% in intracellular S Enteritidis. CONCLUSION: The results demonstrate that S-ßGM contributes to protecting epithelial barrier function in a Caco-2 cell model disrupted by S. Enteritidis.


Subject(s)
Colon/drug effects , Intestinal Mucosa/drug effects , Mannans/pharmacology , Oligosaccharides/pharmacology , Salmonella Infections/physiopathology , Salmonella enteritidis , Tight Junctions/drug effects , Actins/metabolism , Caco-2 Cells , Colon/metabolism , Colon/microbiology , Colon/physiopathology , Dietary Supplements , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Galactose/analogs & derivatives , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Occludin/metabolism , Permeability , Salmonella Infections/microbiology , Zonula Occludens-1 Protein/metabolism
9.
Fish Shellfish Immunol ; 58: 387-396, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666190

ABSTRACT

Salmonella enterica serovar typhimurium (S. typhimurium) is globally distributed and causes massive morbidity and mortality in humans and animals. S. typhimurium carries Salmonella plasmid virulence (spv) locus, which is highly conserved and closely related to bacterial pathogenicity, while its exact role in host immune responses during infection remains to be elucidated. To counteract the invaders, the host has evolved numerous strategies, among which the innate immunity and autophagy act as the first defense. Recently, zebrafish has been universally accepted as a valuable and powerful vertebrate model in analyzing bacteria-host interactions. To investigate whether spv locus enhances the virulence of Salmonella by exerting an effect on the host early defense, zebrafish larvae were employed in this study. LD50 of S. typhimurium to zebrafish larvae and bacterial dissemination were analyzed. Sudan black B and neutral red staining were performed to detect the responses of neutrophils and macrophages to Salmonella infection. Autophagy agonist Torin1 and inhibitor Chloroquine were used to interfere in autophagic flux, and the protein level of Lc3 and p62 were measured by western blotting. Results indicated that spv locus could decrease the LD50 of S. typhimurium to zebrafish larvae, accelerate the reproduction and dissemination of bacteria by inhibiting the function of neutrophils and macrophages. Moreover, spv locus restrained the formation of autophagosomes in the earlier stage of autophagy. These findings suggested the virulence of spv locus involving in suppressing host innate immune responses for the first time, which shed new light on the role of spv operon in Salmonella pathogenicity.


Subject(s)
Bacterial Proteins/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Virulence Factors/genetics , Animals , Bacterial Proteins/metabolism , Disease Models, Animal , Humans , Salmonella Infections/physiopathology , Virulence , Virulence Factors/metabolism , Zebrafish
10.
Fish Shellfish Immunol ; 49: 252-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26723267

ABSTRACT

Salmonella enterica serovar typhimurium (S. typhimurium) is a facultative intracellular pathogen that can cause gastroenteritis and systemic infection in a wide range of hosts. Salmonella plasmid virulence gene spvB is closely related to bacterial virulence in different cells and animal models, and the encoded protein acts as an intracellular toxin required for ADP-ribosyl transferase activity. However, until now there is no report about the pathogenecity of spvB gene on zebrafish. Due to the outstanding advantages of zebrafish in analyzing bacteria-host interactions, a S. typhimurium infected zebrafish model was set up here to study the effect of spvB on autophagy and intestinal pathogenesis in vivo. We found that spvB gene could decrease the LD50 of S. typhimurium, and the strain carrying spvB promoted bacterial proliferation and aggravated the intestinal damage manifested by the narrowed intestines, fallen microvilli, blurred epithelium cell structure and infiltration of inflammatory cells. Results demonstrated the enhanced virulence induced by spvB in zebrafish. In spvB-mutant strain infected zebrafish, the levels of Lc3 turnover and Beclin1 expression increased, and the double-membraned autophagosome structures were observed, suggesting that spvB can inhibit autophagy activity. In summary, our results indicate that S. typhimurium strain containing spvB displays more virulence, triggering an increase in bacterial survival and intestine injuries by suppressing autophagy for the first time. This model provides novel insights into the role of Salmonella plasmid virulence gene in bacterial pathogenesis, and can help to further elucidate the relationship between bacteria and host immune response.


Subject(s)
ADP Ribose Transferases/genetics , ADP Ribose Transferases/metabolism , Autophagy/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Virulence Factors/genetics , Virulence Factors/metabolism , Animals , Disease Models, Animal , Humans , Salmonella Infections/physiopathology , Virulence/genetics , Zebrafish
11.
BMC Infect Dis ; 16: 271, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27286886

ABSTRACT

BACKGROUND: In sub-Saharan Africa, non-typhoidal Salmonella (NTS) can cause bloodstream infections, referred to as invasive non-typhoidal Salmonella disease (iNTS disease); it can occur in outbreaks and is often preceded by malaria. Data from Central Africa is limited. METHODS: Clinical, microbiological and molecular findings of NTS recovered in a blood culture surveillance project (2009-2014) were analyzed. RESULTS: In March-July 2012 there was an epidemic increase in malaria infections in the Oriental Province of the Democratic Republic of the Congo (DRC). In one referral hospital, overall hospital admissions in June 2012 were 2.6 times higher as compared to the same period in the years before and after (336 versus an average of 128 respectively); numbers of malaria cases and blood transfusions were nearly three- and five-fold higher respectively (317 versus 112 and 250 versus 55). Case fatality rates (in-hospital deaths versus all admissions) peaked at 14.6 %. Salmonella Typhimurium and Salmonella Enteritidis together accounted for 88.9 % of pathogens isolated from blood cultures collected during an outreach visit to the affected districts in June 2012. Children infected with Salmonella Enteritidis (33 patient files available) tended to be co-infected with Plasmodium falciparum more often than children infected with Salmonella Typhimurium (40 patients files available) (81.8 % versus 62.5 %). Through the microbiological surveillance project (May 2009-May 2014) 113 unique NTS isolates were collected (28.5 % (113/396) of pathogens); most (95.3 %) were recovered from children < 15 years. Salmonella Typhimurium (n = 54) and Salmonella Enteritidis (n = 56) accounted for 47.8 % and of 49.6 % NTS isolates respectively. Multilocus variable-number tandem-repeat analysis (MLVA) revealed more heterogeneity for Salmonella Typhimurium than for Salmonella Enteritidis. Most (82/96, 85.4 %) NTS isolates that were available for antibiotic susceptibility testing were multidrug resistant. All isolates were susceptible to ceftriaxone and azithromycin. CONCLUSION: During the peak of an epidemic increase in malaria in the DRC in 2012, a high proportion of multidrug resistant Salmonella Typhimurium and Salmonella Enteritidis were isolated from blood cultures. Overall, the two serovars showed subtle differences in clinical presentation and genetic diversity.


Subject(s)
Bacteremia/epidemiology , Coinfection/epidemiology , Malaria, Falciparum/epidemiology , Salmonella Infections/epidemiology , Adolescent , Adult , Anti-Bacterial Agents/therapeutic use , Asian People , Azithromycin/therapeutic use , Bacteremia/drug therapy , Bacteremia/microbiology , Bacteremia/physiopathology , Ceftriaxone/therapeutic use , Child , Child, Preschool , Democratic Republic of the Congo/epidemiology , Disease Outbreaks , Drug Resistance, Multiple, Bacterial , Female , Hospitalization , Humans , Infant , Infant, Newborn , Malaria/epidemiology , Male , Salmonella Infections/drug therapy , Salmonella Infections/microbiology , Salmonella Infections/physiopathology , Salmonella enteritidis/genetics , Salmonella enteritidis/isolation & purification , Salmonella enteritidis/physiology , Salmonella typhimurium/genetics , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/physiology , Serogroup , Tandem Repeat Sequences
12.
Nature ; 468(7327): 1119-23, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21068722

ABSTRACT

Interaction of pathogens with cells of the immune system results in activation of inflammatory gene expression. This response, although vital for immune defence, is frequently deleterious to the host due to the exaggerated production of inflammatory proteins. The scope of inflammatory responses reflects the activation state of signalling proteins upstream of inflammatory genes as well as signal-induced assembly of nuclear chromatin complexes that support mRNA expression. Recognition of post-translationally modified histones by nuclear proteins that initiate mRNA transcription and support mRNA elongation is a critical step in the regulation of gene expression. Here we present a novel pharmacological approach that targets inflammatory gene expression by interfering with the recognition of acetylated histones by the bromodomain and extra terminal domain (BET) family of proteins. We describe a synthetic compound (I-BET) that by 'mimicking' acetylated histones disrupts chromatin complexes responsible for the expression of key inflammatory genes in activated macrophages, and confers protection against lipopolysaccharide-induced endotoxic shock and bacteria-induced sepsis. Our findings suggest that synthetic compounds specifically targeting proteins that recognize post-translationally modified histones can serve as a new generation of immunomodulatory drugs.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammation , Macrophages/drug effects , Acetylation/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Benzodiazepines , Cells, Cultured , Epigenomics , Genome-Wide Association Study , Heterocyclic Compounds, 4 or More Rings/chemistry , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Inflammation/drug therapy , Inflammation/prevention & control , Kaplan-Meier Estimate , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Salmonella Infections/drug therapy , Salmonella Infections/immunology , Salmonella Infections/physiopathology , Salmonella Infections/prevention & control , Salmonella typhimurium , Sepsis/drug therapy , Sepsis/prevention & control , Shock, Septic/drug therapy , Shock, Septic/prevention & control
13.
Immunol Rev ; 245(1): 56-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22168414

ABSTRACT

The mammalian intestine is colonized by a dense microbial community, the microbiota. Homeostatic and symbiotic interactions facilitate the peaceful co-existence between the microbiota and the host, and inhibit colonization by most incoming pathogens ('colonization resistance'). However, if pathogenic intruders overcome colonization resistance, a fierce, innate inflammatory defense can be mounted within hours, the adaptive arm of the immune system is initiated, and the pathogen is fought back. The molecular nature of the homeostatic interactions, the pathogen's ability to overcome colonization resistance, and the triggering of native and adaptive mucosal immune responses are still poorly understood. To study these mechanisms, the streptomycin mouse model for Salmonella diarrhea is of great value. Here, we review how S. Typhimurium triggers mucosal immune responses by active (virulence factor elicited) and passive (MyD88-dependent) mechanisms and introduce the S. Typhimurium mutants available for focusing on either response. Interestingly, mucosal defense turns out to be a double-edged sword, limiting pathogen burdens in the gut tissue but enhancing pathogen growth in the gut lumen. This model allows not only studying the molecular pathogenesis of Salmonella diarrhea but also is ideally suited for analyzing innate defenses, microbe handling by mucosal phagocytes, adaptive secretory immunoglobulin A responses, probing microbiota function, and homeostatic microbiota-host interactions. Finally, we discuss the general need for defined assay conditions when using animal models for enteric infections and the central importance of littermate controls.


Subject(s)
Diarrhea/immunology , Diarrhea/microbiology , Host-Pathogen Interactions/immunology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella/immunology , Virulence Factors/immunology , Animals , Diarrhea/etiology , Diarrhea/physiopathology , Disease Models, Animal , Humans , Immunity, Mucosal , Metagenome/genetics , Metagenome/immunology , Mice , Salmonella/genetics , Salmonella Infections/complications , Salmonella Infections/physiopathology , Streptomycin/administration & dosage , Streptomycin/adverse effects , Virulence
15.
Infect Immun ; 82(1): 333-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166960

ABSTRACT

Herein we report an important role for the ferric uptake regulator (Fur) in the resistance of Salmonella enterica serovar Typhimurium to the reactive nitrogen species produced by inducible nitric oxide (NO) synthase in an NRAMP1(r) murine model of acute systemic infection. The expression of fur protected Salmonella grown under normoxic and hypoxic conditions against the bacteriostatic activity of NO. The hypersusceptibility of fur-deficient Salmonella to the cytotoxic actions of NO coincides with a marked repression of respiratory activity and the reduced ability of the bacteria to detoxify NO. A fur mutant Salmonella strain contained reduced levels of the terminal quinol oxidases of the electron transport chain. Addition of the heme precursor δ-aminolevulinic acid restored the cytochrome content, respiratory activity, NO consumption, and wild-type growth in bacteria undergoing nitrosative stress. The innate antinitrosative defenses regulated by Fur added to the adaptive response associated with the NO-detoxifying activity of the flavohemoprotein Hmp. Our investigations indicate that, in addition to playing a critical role in iron homeostasis, Fur is an important antinitrosative determinant of Salmonella pathogenesis.


Subject(s)
Bacterial Proteins/physiology , Repressor Proteins/physiology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Analysis of Variance , Animals , Bacterial Proteins/immunology , Disease Models, Animal , Mice , Nitric Oxide Synthase Type II , Oxidative Stress/physiology , Repressor Proteins/deficiency , Repressor Proteins/immunology , Salmonella Infections/physiopathology , Salmonella typhimurium/immunology , Stress, Physiological/physiology
16.
J Cell Sci ; 125(Pt 8): 1991-2003, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22328521

ABSTRACT

A balance between endocytosis and membrane recycling regulates the composition and dynamics of the plasma membrane. Internalization and recycling of cholesterol- and sphingolipid-enriched lipid rafts is an actin-dependent process that is mediated by a specialized Arf6-dependent recycling pathway. Here, we identify myosin1c (Myo1c) as the first motor protein that drives the formation of recycling tubules emanating from the perinuclear recycling compartment. We demonstrate that the single-headed Myo1c is a lipid-raft-associated motor protein that is specifically involved in recycling of lipid-raft-associated glycosylphosphatidylinositol (GPI)-linked cargo proteins and their delivery to the cell surface. Whereas Myo1c overexpression increases the levels of these raft proteins at the cell surface, in cells depleted of Myo1c function through RNA interference or overexpression of a dominant-negative mutant, these tubular transport carriers of the recycling pathway are lost and GPI-linked raft markers are trapped in the perinuclear recycling compartment. Intriguingly, Myo1c only selectively promotes delivery of lipid raft membranes back to the cell surface and is not required for recycling of cargo, such as the transferrin receptor, which is mediated by parallel pathways. The profound defect in lipid raft trafficking in Myo1c-knockdown cells has a dramatic impact on cell spreading, cell migration and cholesterol-dependent Salmonella invasion; processes that require lipid raft transport to the cell surface to deliver signaling components and the extra membrane essential for cell surface expansion and remodeling. Thus, Myo1c plays a crucial role in the recycling of lipid raft membrane and proteins that regulate plasma membrane plasticity, cell motility and pathogen entry.


Subject(s)
Cell Movement , Lipid Metabolism , Membrane Lipids/metabolism , Myosin Type I/metabolism , Salmonella Infections/metabolism , Salmonella Infections/physiopathology , Salmonella typhimurium/physiology , Biological Transport , Cholesterol/metabolism , Exocytosis , HeLa Cells , Humans , Myosin Type I/genetics , Salmonella Infections/genetics , Salmonella Infections/microbiology
17.
J Immunol ; 189(9): 4266-74, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22993205

ABSTRACT

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4(+) thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Subject(s)
Recovery of Function/immunology , Salmonella Infections/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Animals , Atrophy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Differentiation/immunology , Cell Movement/immunology , Mice , Salmonella Infections/pathology , Salmonella Infections/physiopathology , Salmonella typhimurium/immunology , Thymus Gland/microbiology
18.
J Immunol ; 185(10): 6198-204, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20952675

ABSTRACT

Systemic Salmonella infection commonly induces prolonged splenomegaly in murine or human hosts. Although this increase in splenic cellularity is often assumed to be due to the recruitment and expansion of leukocytes, the actual cause of splenomegaly remains unclear. We monitored spleen cell populations during Salmonella infection and found that the most prominent increase is found in the erythroid compartment. At the peak of infection, the majority of spleen cells are immature CD71(-)Ter119(+) reticulocytes, indicating that massive erythropoiesis occurs in response to Salmonella infection. Indeed, this increase in RBC precursors corresponded with marked elevation of serum erythropoietin (EPO). Furthermore, the increase in RBC precursors and EPO production required innate immune signaling mediated by Myd88/TRIF. Neutralization of EPO substantially reduced the immature RBC population in the spleen and allowed a modest increase in host control of infection. These data indicate that early innate immunity to Salmonella initiates marked splenic erythropoiesis and may hinder bacterial clearance.


Subject(s)
Erythropoiesis/immunology , Immunity, Innate/immunology , Salmonella Infections/immunology , Splenomegaly/immunology , Animals , Erythropoietin/blood , Flow Cytometry , Immunoassay , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reticulocytes/immunology , Salmonella Infections/blood , Salmonella Infections/physiopathology , Splenomegaly/microbiology , Splenomegaly/parasitology
19.
Cell Mol Life Sci ; 68(22): 3687-97, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21984608

ABSTRACT

Acute gastroenteritis caused by Salmonella enterica serovar typhimurium is a significant public health problem. This pathogen has very sophisticated molecular machinery encoded by the two pathogenicity islands, namely Salmonella Pathogenicity Island 1 and 2 (SPI-1 and SPI-2). Remarkably, both SPI-1 and SPI-2 are very tightly regulated in terms of timing of expression and spatial localization of the encoded effectors during the infection process within the host cell. This regulation is governed at several levels, including transcription and translation, and by post-translational modifications. In the context of a finely tuned regulatory system, we will highlight how these effector proteins co-opt host signaling pathways that control the ability of the organism to infect and survive within the host, as well as elicit host pro-inflammatory responses.


Subject(s)
Bacterial Proteins/immunology , Host-Pathogen Interactions/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Genomic Islands , Humans , Immunity, Innate/immunology , Protein Processing, Post-Translational , Salmonella Infections/physiopathology , Salmonella typhimurium/genetics , Signal Transduction/physiology , Vacuoles/metabolism , Vacuoles/microbiology
20.
BMC Public Health ; 12: 310, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22537320

ABSTRACT

BACKGROUND: Increases in the number of salmonellosis cases due to Salmonella Enteritidis (SE) in 2010 and 2011 prompted a public health investigation in Ontario, Canada. In this report, we describe the current epidemiology of travel-related (TR) SE, compare demographics, symptoms and phage types (PTs) of TR and domestically-acquired (DA) cases, and estimate the odds of acquiring SE by region of the world visited. METHODS: All incident cases of culture confirmed SE in Ontario obtained from isolates and specimens submitted to public health laboratories were included in this study. Demographic and illness characteristics of TR and DA cases were compared. A national travel survey was used to provide estimates for the number of travellers to various destinations to approximate rates of SE in travellers. Multivariate logistic regression was used to estimate the odds of acquiring SE when travelling to various world regions. RESULTS: Overall, 51.9% of SE cases were TR during the study period. This ranged from 35.7% TR cases in the summer travel period to 65.1% TR cases in the winter travel period. Compared to DA cases, TR cases were older and were less likely to seek hospital care. For Ontario travellers, the adjusted odds of acquiring SE was the highest for the Caribbean (OR 37.29, 95% CI 17.87-77.82) when compared to Europe. Certain PTs were more commonly associated with travel (e.g., 1, 4, 5b, 7a, Atypical) than with domestic infection. Of the TR cases, 88.9% were associated with travel to the Caribbean and Mexico region, of whom 90.1% reported staying on a resort. Within this region, there were distinct associations between PTs and countries. CONCLUSIONS: There is a large burden of TR illness from SE in Ontario. Accurate classification of cases by travel history is important to better understand the source of infections. The findings emphasize the need to make travellers, especially to the Caribbean, and health professionals who provide advice to travellers, aware of this risk. The findings may be generalized to other jurisdictions with travel behaviours in their residents similar to Ontario residents.


Subject(s)
Salmonella Infections/epidemiology , Salmonella enteritidis , Travel , Adolescent , Adult , Antigua and Barbuda , Chi-Square Distribution , Child , Child, Preschool , Cuba , Disease Transmission, Infectious , Dominican Republic , Female , Humans , Infant , Infant, Newborn , Jamaica , Logistic Models , Male , Middle Aged , Ontario/epidemiology , Patient Acceptance of Health Care , Salmonella Infections/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL