Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2813: 189-204, 2024.
Article in English | MEDLINE | ID: mdl-38888779

ABSTRACT

Classic in vitro coculture assays of pathogens with host cells have contributed significantly to our understanding of the intracellular lifestyle of several pathogens. Coculture assays with pathogens and eukaryotic cells can be analyzed through various techniques including plating for colony-forming units (CFU), confocal microscopy, and flow cytometry. However, findings from in vitro assays require validation in an in vivo model. Several physiological conditions can influence host-pathogen interactions, which cannot easily be mimicked in vitro. Intravital microscopy (IVM) is emerging as a powerful tool for studying host-pathogen interactions by enabling in vivo imaging of living organisms. As a result, IVM has significantly enhanced the understanding of infection mediated by diverse pathogens. The versatility of IVM has also allowed for the imaging of various organs as sites of local infection. This chapter specifically focuses on IVM conducted on the lung for elucidating pulmonary immune response, primarily involving alveolar macrophages, to pathogens. Additionally, in this chapter we outline the protocol for lung IVM that utilizes a thoracic suction window to stabilize the lung for acquiring stable images.


Subject(s)
Cell Tracking , Intravital Microscopy , Macrophages, Alveolar , Macrophages, Alveolar/cytology , Intravital Microscopy/methods , Animals , Cell Tracking/methods , Mice , Lung/cytology , Host-Pathogen Interactions
2.
Nature ; 614(7948): 530-538, 2023 02.
Article in English | MEDLINE | ID: mdl-36599368

ABSTRACT

Resident-tissue macrophages (RTMs) arise from embryonic precursors1,2, yet the developmental signals that shape their longevity remain largely unknown. Here we demonstrate in mice genetically deficient in 12-lipoxygenase and 15-lipoxygenase (Alox15-/- mice) that neonatal neutrophil-derived 12-HETE is required for self-renewal and maintenance of alveolar macrophages (AMs) during lung development. Although the seeding and differentiation of AM progenitors remained intact, the absence of 12-HETE led to a significant reduction in AMs in adult lungs and enhanced senescence owing to increased prostaglandin E2 production. A compromised AM compartment resulted in increased susceptibility to acute lung injury induced by lipopolysaccharide and to pulmonary infections with influenza A virus or SARS-CoV-2. Our results highlight the complexity of prenatal RTM programming and reveal their dependency on in trans eicosanoid production by neutrophils for lifelong self-renewal.


Subject(s)
12-Hydroxy-5,8,10,14-eicosatetraenoic Acid , Cell Self Renewal , Macrophages, Alveolar , Neutrophils , Animals , Mice , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Acute Lung Injury , Animals, Newborn , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 15-Lipoxygenase/deficiency , COVID-19 , Influenza A virus , Lipopolysaccharides , Lung/cytology , Lung/virology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Neutrophils/metabolism , Orthomyxoviridae Infections , Prostaglandins E , SARS-CoV-2 , Disease Susceptibility
3.
Front Immunol ; 13: 1029085, 2022.
Article in English | MEDLINE | ID: mdl-36532044

ABSTRACT

Resident macrophages play a unique role in the maintenance of tissue function. As phagocytes, they are an essential first line defenders against pathogens and much of the initial characterization of these cells was focused on their interaction with viral and bacterial pathogens. However, these cells are increasingly recognized as contributing to more than just host defense. Through cytokine production, receptor engagement and gap junction communication resident macrophages tune tissue inflammatory tone, influence adaptive immune cell phenotype and regulate tissue structure and function. This review highlights resident macrophages in the liver and lung as they hold unique roles in the maintenance of the interface between the circulatory system and the external environment. As such, we detail the developmental origin of these cells, their contribution to host defense and the array of tools these cells use to regulate tissue homeostasis.


Subject(s)
Liver , Macrophages , Lung , Phagocytes , Homeostasis
4.
WIREs Mech Dis ; 14(5): e1567, 2022 09.
Article in English | MEDLINE | ID: mdl-35674186

ABSTRACT

In a world filled with microbes, some posing a threat to our body, our immune system is key to living a healthy life. The innate immune system is made of various cell types that act to guard our bodies. Unlike the adaptive immune system that has a specific response, our innate immune system encompasses cells that elicit unspecific immune responses, triggered whenever the right signals are detected. Our understanding of immunity started with the concept of our immune system only responding to "nonself" like the pathogens that invade our body. However, over the past few decades, we have learned that the immune system is more than an on/off switch that recognizes nonself. The innate immune system regularly patrols our bodies for pathogens and tissue damage. Our innate immune system not only seeks to resolve infection but also repair tissue injury, through phagocytosing debris and initiating the release of growth factors. Recently, we are starting to see that it is not just recognizing danger, our innate immune system plays a crucial role in repair. Innate immune cells phenotypically change during repair. In the context of severe injury or trauma, our innate immune system is modified quite drastically to help repair, resulting in reduced infection control. Moreover, these changes in immune cell function can be modified by sex as a biological variable. From past to present, in this overview, we provide a summary of the innate immune cells and pathways in infection and tissue repair. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology.


Subject(s)
Immune System , Immunity, Innate , Immunity, Innate/physiology , Phagocytosis
5.
Int J Biochem Cell Biol ; 134: 105966, 2021 05.
Article in English | MEDLINE | ID: mdl-33677070

ABSTRACT

As we learn more about chronic lung diseases, we are seeing that an unbalanced immune system plays a key role in disease pathogenesis. Innate immune cells, particularly tissue-resident macrophages, are important navigators of immunity, both during infection and in non-communicable lung disease. In the lung, alveolar macrophages are considered some of the most critical and diverse immune cells, yet despite an array of studies over the years, alveolar macrophages remain poorly understood. In this review, we highlight the importance of alveolar macrophages in health and disease, and discuss how proteomics can be used to elucidate mechanistic information and identify potential targets for therapy development.


Subject(s)
Inflammation/immunology , Lung Diseases/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Proteome/immunology , Proteomics/methods , Animals , Biomarkers/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Lung/pathology , Lung Diseases/metabolism , Lung Diseases/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Proteome/metabolism
6.
Sci Adv ; 7(10)2021 03.
Article in English | MEDLINE | ID: mdl-33674305

ABSTRACT

The underlying mechanisms contributing to injury-induced infection susceptibility remain poorly understood. Here, we describe a rapid increase in neutrophil cell numbers in the lungs following induction of thermal injury. These neutrophils expressed elevated levels of programmed death ligand 1 (PD-L1) and exhibited altered gene expression profiles indicative of a reparative population. Upon injury, neutrophils migrate from the bone marrow to the skin but transiently arrest in the lung vasculature. Arrested neutrophils interact with programmed cell death protein 1 (PD-1) on lung endothelial cells. A period of susceptibility to infection is linked to PD-L1+ neutrophil accumulation in the lung. Systemic treatment of injured animals with an anti-PD-L1 antibody prevented neutrophil accumulation in the lung and reduced susceptibility to infection but augmented skin healing, resulting in increased epidermal growth. This work provides evidence that injury promotes changes to neutrophils that are important for wound healing but contribute to infection susceptibility.

7.
Cell ; 183(1): 110-125.e11, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32888431

ABSTRACT

During respiration, humans breathe in more than 10,000 liters of non-sterile air daily, allowing some pathogens access to alveoli. Interestingly, alveoli outnumber alveolar macrophages (AMs), which favors alveoli devoid of AMs. If AMs, like most tissue macrophages, are sessile, then this numerical advantage would be exploited by pathogens unless neutrophils from the blood stream intervened. However, this would translate to omnipresent persistent inflammation. Developing in vivo real-time intravital imaging of alveoli revealed AMs crawling in and between alveoli using the pores of Kohn. Importantly, these macrophages sensed, chemotaxed, and, with high efficiency, phagocytosed inhaled bacterial pathogens such as P. aeruginosa and S. aureus, cloaking the bacteria from neutrophils. Impairing AM chemotaxis toward bacteria induced superfluous neutrophil recruitment, leading to inappropriate inflammation and injury. In a disease context, influenza A virus infection impaired AM crawling via the type II interferon signaling pathway, and this greatly increased secondary bacterial co-infection.


Subject(s)
Bacteria/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Animals , Female , Homeostasis , Humans , Lung/immunology , Lung/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Neutrophils/immunology , Phagocytosis/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Pulmonary Alveoli , Signal Transduction , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
8.
Cell ; 178(5): 1205-1221.e17, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442408

ABSTRACT

A hallmark feature of inflammation is the orchestrated recruitment of neutrophils from the bloodstream into inflamed tissue. Although selectins and integrins mediate recruitment in many tissues, they have a minimal role in the lungs and liver. Exploiting an unbiased in vivo functional screen, we identified a lung and liver homing peptide that functionally abrogates neutrophil recruitment to these organs. Using biochemical, genetic, and confocal intravital imaging approaches, we identified dipeptidase-1 (DPEP1) as the target and established its role as a physical adhesion receptor for neutrophil sequestration independent of its enzymatic activity. Importantly, genetic ablation or functional peptide blocking of DPEP1 significantly reduced neutrophil recruitment to the lungs and liver and provided improved survival in models of endotoxemia. Our data establish DPEP1 as a major adhesion receptor on the lung and liver endothelium and identify a therapeutic target for neutrophil-driven inflammatory diseases of the lungs.


Subject(s)
Dipeptidases/metabolism , Neutrophils/physiology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Animals , Cilastatin/pharmacology , Cilastatin/therapeutic use , Dipeptidases/antagonists & inhibitors , Dipeptidases/genetics , Disease Models, Animal , Endotoxemia/mortality , Endotoxemia/pathology , Endotoxemia/prevention & control , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/immunology , Liver/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Neutrophil Infiltration/drug effects , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Survival Rate
9.
J Leukoc Biol ; 105(6): 1083-1084, 2019 06.
Article in English | MEDLINE | ID: mdl-31087694

ABSTRACT

Discussion on neutrophil extracellular traps (NETs) clearing inflammation from the closed eye, argues the role of NETs as an anti-inflammatory component of immunity.


Subject(s)
Extracellular Traps , Anti-Inflammatory Agents , Humans , Inflammation , Neutrophils
10.
Cell Host Microbe ; 25(4): 526-536.e4, 2019 Apr 10.
Article in English | MEDLINE | ID: mdl-30930127

ABSTRACT

Bacterial biofilm infections are difficult to eradicate because of antibiotic insusceptibility and high recurrence rates. Biofilm formation by Pseudomonas aeruginosa, a leading cause of bacterial keratitis, is facilitated by the bacterial Psl exopolysaccharide and associated with heightened virulence. Using intravital microscopy, we observed that neutrophilic recruitment to corneal infections limits P. aeruginosa biofilms to the outer eye surface, preventing bacterial dissemination. Neutrophils moved to the base of forming biofilms, where they underwent neutrophil extracellular trap formation (NETosis) in response to high expression of the bacterial type-3 secretion system (T3SS). NETs formed a barrier "dead zone," confining bacteria to the external corneal environment and inhibiting bacterial dissemination into the brain. Once formed, ocular biofilms were resistant to antibiotics and neutrophil killing, advancing eye pathology. However, blocking both Psl and T3SS together with antibiotic treatment broke down the biofilm and reversed keratitis, suggesting future therapeutic strategies for this intractable infection.


Subject(s)
Biofilms/growth & development , Cornea/microbiology , Extracellular Traps/metabolism , Meningoencephalitis/prevention & control , Neutrophils/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Disease Models, Animal , Mice , Pseudomonas Infections/complications , Pseudomonas aeruginosa/growth & development
11.
J Leukoc Biol ; 106(2): 323-335, 2019 08.
Article in English | MEDLINE | ID: mdl-30776153

ABSTRACT

It has long been appreciated that understanding the interactions between the host and the pathogens that make us sick is critical for the prevention and treatment of disease. As antibiotics become increasingly ineffective, targeting the host and specific bacterial evasion mechanisms are becoming novel therapeutic approaches. The technology used to understand host-pathogen interactions has dramatically advanced over the last century. We have moved away from using simple in vitro assays focused on single-cell events to technologies that allow us to observe complex multicellular interactions in real time in live animals. Specifically, intravital microscopy (IVM) has improved our understanding of infection, from viral to bacterial to parasitic, and how the host immune system responds to these infections. Yet, at the same time it has allowed us to appreciate just how complex these interactions are and that current experimental models still have a number of limitations. In this review, we will discuss the advances in vivo IVM has brought to the study of host-pathogen interactions, focusing primarily on bacterial infections and innate immunity.


Subject(s)
Communicable Diseases/etiology , Host-Pathogen Interactions/immunology , Immunity , Animals , Communicable Diseases/diagnosis , Communicable Diseases/metabolism , Diagnostic Imaging/methods , Disease Susceptibility , Humans , Immunity, Innate , Intravital Microscopy , Organ Specificity , Severity of Illness Index
12.
Cell Host Microbe ; 24(2): 271-284.e3, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30033122

ABSTRACT

During sepsis, small blood vessels can become occluded by large platelet aggregates of poorly understood etiology. During Staphylococcal aureus infection, sepsis severity is linked to the bacterial α-toxin (α-hemolysin, AT) through unclear mechanisms. In this study, we visualized intravascular events in the microcirculation and found that intravenous AT injection induces rapid platelet aggregation, forming dynamic micro-thrombi in the microcirculation. These aggregates are retained in the liver sinusoids and kidney glomeruli, causing multi-organ dysfunction. Acute staphylococcal infection results in sequestration of most bacteria by liver macrophages. Platelets are initially recruited to these macrophages and help eradicate S. aureus. However, at later time points, AT causes aberrant and damaging thrombosis throughout the liver. Treatment with an AT neutralizing antibody (MEDI4893∗) prevents platelet aggregation and subsequent liver damage, without affecting the initial and beneficial platelet recruitment. Thus, AT neutralization may represent a promising approach to combat staphylococcal-induced intravascular coagulation and organ dysfunction.


Subject(s)
Bacteremia/physiopathology , Bacterial Toxins/toxicity , Hemolysin Proteins/toxicity , Liver/pathology , Platelet Aggregation/drug effects , Staphylococcal Infections/physiopathology , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/immunology , Broadly Neutralizing Antibodies , Hemolysin Proteins/immunology , Host-Pathogen Interactions/physiology , Humans , Intravital Microscopy/methods , Liver/drug effects , Liver/microbiology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Platelet Aggregation/physiology , Staphylococcus aureus/pathogenicity
13.
Science ; 358(6359): 111-116, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28983053

ABSTRACT

Neutrophils have been implicated as harmful cells in a variety of inappropriate inflammatory conditions where they injure the host, leading to the death of the neutrophils and their subsequent phagocytosis by monocytes and macrophages. Here we show that in a fully repairing sterile thermal hepatic injury, neutrophils also penetrate the injury site and perform the critical tasks of dismantling injured vessels and creating channels for new vascular regrowth. Upon completion of these tasks, they neither die at the injury site nor are phagocytosed. Instead, many of these neutrophils reenter the vasculature and have a preprogrammed journey that entails a sojourn in the lungs to up-regulate CXCR4 (C-X-C motif chemokine receptor 4) before entering the bone marrow, where they undergo apoptosis.


Subject(s)
Neovascularization, Physiologic/immunology , Neutrophils/immunology , Wound Healing/immunology , Animals , Apoptosis/immunology , Bone Marrow/immunology , Cell Movement/immunology , Liver/blood supply , Liver/injuries , Lung/immunology , Mice , Mice, Inbred C57BL , Receptors, CXCR4/metabolism , Up-Regulation
14.
J Clin Invest ; 127(6): 2249-2261, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28463232

ABSTRACT

Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Bispecific , Antigens, Bacterial/immunology , Bacterial Load , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Complement System Proteins/metabolism , Drug Evaluation, Preclinical , Female , Kupffer Cells/microbiology , Lung/blood supply , Lung/microbiology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Microvessels/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Pore Forming Cytotoxic Proteins/immunology , Pseudomonas Infections/immunology , Receptors, Fc/metabolism
15.
Cell Rep ; 16(12): 3260-3272, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653688

ABSTRACT

iNKT cells are a subset of innate T cells that recognize glycolipids presented on CD1d molecules and protect against bacterial infections, including S. pneumoniae. Using lung intravital imaging, we examined the behavior and mechanism of pulmonary iNKT cell activation in response to the specific iNKT cell ligand α-galactosylceramide or S. pneumoniae infection. In untreated mice, the major fraction of iNKT cells resided in the vasculature, but a small critical population resided in the extravascular space in proximity to monocyte-derived DCs. Administration of either α-GalCer or S. pneumoniae induced CD1d-dependent rapid recruitment of neutrophils out of the vasculature. The neutrophils guided iNKT cells from the lung vasculature via CCL17. Depletion of monocyte-derived DCs abrogated both the neutrophil and subsequent iNKT cell extravasation. Moreover, impairing iNKT cell recruitment by blocking CCL17 increased susceptibility to S. pneumoniae infection, suggesting a critical role for the influx of iNKT cells in host defense.


Subject(s)
Dendritic Cells/immunology , Inflammation/immunology , Natural Killer T-Cells/immunology , Neutrophils/immunology , Pneumococcal Infections/immunology , Animals , Chemotaxis, Leukocyte/immunology , Lung/immunology , Lung/microbiology , Mice , Mice, Transgenic , Monocytes/cytology , Monocytes/immunology
16.
Nat Commun ; 6: 6673, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25809117

ABSTRACT

Neutrophil extracellular traps (NETs) composed of DNA decorated with histones and proteases trap and kill bacteria but also injure host tissue. Here we show that during a bloodstream infection with methicillin-resistant Staphylococcus aureus, the majority of bacteria are sequestered immediately by hepatic Kupffer cells, resulting in transient increases in liver enzymes, focal ischaemic areas and a robust neutrophil infiltration into the liver. The neutrophils release NETs into the liver vasculature, which remain anchored to the vascular wall via von Willebrand factor and reveal significant neutrophil elastase (NE) proteolytic activity. Importantly, DNase although very effective at DNA removal, and somewhat effective at inhibiting NE proteolytic activity, fails to remove the majority of histones from the vessel wall and only partly reduces injury. By contrast, inhibition of NET production as modelled by PAD4-deficiency, or prevention of NET formation and proteolytic activity as modelled in NE(-/-) mice prevent collateral host tissue damage.


Subject(s)
Bacteremia/immunology , Extracellular Traps/immunology , Hepatic Artery/immunology , Hepatic Veins/immunology , Leukocyte Elastase/genetics , Liver/immunology , Staphylococcal Infections/immunology , Animals , Bacteremia/metabolism , Deoxyribonucleases/metabolism , Hepatic Artery/metabolism , Hepatic Veins/metabolism , Histones/metabolism , Hydrolases/genetics , Kupffer Cells/immunology , Leukocyte Elastase/metabolism , Liver/blood supply , Liver/enzymology , Liver/metabolism , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Knockout , Neutrophil Infiltration , Protein-Arginine Deiminase Type 4 , Staphylococcal Infections/metabolism , von Willebrand Factor/metabolism
18.
Microbes Infect ; 15(10-11): 738-44, 2013.
Article in English | MEDLINE | ID: mdl-23747681

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) and enteropathogenic E. coli (EPEC) are bacterial pathogens that cause severe illnesses in humans. Citrobacter rodentium is a related mouse pathogen that serves as a small animal model for EPEC and EHEC infections. EPEC, EHEC and C. rodentium translocate bacterial virulence proteins directly into host intestinal cells via a type III secretion system (T3SS). Non-LEE-encoded effector A (NleA) is a T3SS effector that is common to EPEC, EHEC and C. rodentium. NleA interacts with and inhibits the mammalian COPII complex, impairing cellular secretion; this interaction is required for bacterial virulence. Although diarrhea is a hallmark of EPEC, EHEC and C. rodentium infections, the underlying mechanisms are not well characterized. One of the essential functions of the intestine is to maintain a barrier between the lumen and submucosa. Tight junctions seal the space between adjacent epithelial cells creating this barrier. Consequently, it is thought that the disruption of intestinal epithelial tight junctions by EPEC, EHEC, and C. rodentium could result in a loss of barrier function. In this study, we demonstrate that NleA mediated COPII inhibition is required for EPEC- and C. rodentium-mediated disruption of tight junction proteins and increases in fecal water content.


Subject(s)
COP-Coated Vesicles/metabolism , Citrobacter rodentium/physiology , Diarrhea/microbiology , Enteropathogenic Escherichia coli/physiology , Epithelial Cells/physiology , Escherichia coli Proteins/metabolism , Tight Junctions , Virulence Factors/metabolism , Animals , Diarrhea/pathology , Disease Models, Animal , Epithelial Cells/microbiology , Host-Pathogen Interactions , Mice , Mice, Inbred C3H
19.
Nat Commun ; 4: 1898, 2013.
Article in English | MEDLINE | ID: mdl-23695692

ABSTRACT

Citrobacter rodentium is a natural mouse pathogen widely used as a model for enteropathogenic and enterohemorrhagic Escherichia coli infections in humans. While C. rodentium causes self-limiting colitis in most inbred mouse strains, it induces fatal diarrhoea in susceptible strains. The physiological pathways as well as the genetic determinants leading to susceptibility have remained largely uncharacterized. Here we use a forward genetic approach to identify the R-spondin2 gene as a major determinant of susceptibility to C. rodentium infection. Robust induction of R-spondin2 expression during infection in susceptible mouse strains causes a potent Wnt-mediated proliferative response of colonic crypt cells, leading to the generation of an immature and poorly differentiated colonic epithelium with deficiencies in ion-transport components. Our data demonstrate a previously unknown role of R-spondins and Wnt signalling in susceptibility to infectious diarrhoea and identify R-spondin2 as a key molecular link between infection and intestinal homoeostasis.


Subject(s)
Citrobacter rodentium/physiology , Diarrhea/metabolism , Diarrhea/microbiology , Disease Susceptibility/microbiology , Enterobacteriaceae Infections/metabolism , Signal Transduction , Thrombospondins/metabolism , Animals , Cell Differentiation , Chromosome Mapping , Cloning, Molecular , Colon/microbiology , Colon/pathology , Diarrhea/pathology , Disease Susceptibility/metabolism , Disease Susceptibility/pathology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Genetic Association Studies , Genetic Loci/genetics , Humans , Hyperplasia , Mice , Mice, Inbred Strains , Microvilli/microbiology , Microvilli/pathology , Models, Biological , Stromal Cells/metabolism , Stromal Cells/microbiology , Stromal Cells/pathology , Survival Analysis , Wnt Signaling Pathway
20.
Appl Environ Microbiol ; 79(12): 3892-6, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23584785

ABSTRACT

This study identified and characterized enteropathogenic Escherichia coli (EPEC) in the Canadian food supply. Eighteen of 450 E. coli isolates from food animal sources were identified as atypical EPEC (aEPEC). Several of the aEPEC isolates identified in this study possessed multiple virulence genes, exhibited adherence and attaching and effacing (A/E) lesion formation, disrupted tight junctions, and were coclassified with the extraintestinal pathogenic E. coli (ExPEC) and enterotoxigenic E. coli (ETEC) pathotypes.


Subject(s)
Cattle Diseases/epidemiology , Chickens , Enteropathogenic Escherichia coli/genetics , Escherichia coli Infections/veterinary , Meat/microbiology , Poultry Diseases/epidemiology , Swine Diseases/epidemiology , Abattoirs/standards , Animals , Blotting, Western/veterinary , Canada/epidemiology , Cattle , Cattle Diseases/microbiology , Cluster Analysis , Drug Resistance, Microbial/genetics , Escherichia coli Infections/epidemiology , Food Supply/standards , Microscopy, Fluorescence , Polymerase Chain Reaction/veterinary , Poultry Diseases/microbiology , Swine , Swine Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...