Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Gut Microbes ; 14(1): 2143224, 2022.
Article in English | MEDLINE | ID: mdl-36476073

ABSTRACT

The diarrheagenic pathogen enteropathogenic Escherichia coli is responsible for significant childhood mortality and morbidity. EPEC and related attaching-and-effacing (A/E) pathogens use a type III secretion system to hierarchically deliver effector proteins into host cells and manipulate epithelial structure and function. Subversion of host mitochondrial biology is a key aspect of A/E pathogen virulence strategy, but the mechanisms remain poorly defined. We demonstrate that the early-secreted effector EspZ and the late-secreted effector EspH have contrasting effects on host mitochondrial structure and function. EspZ interacts with FIS1, a protein that induces mitochondrial fragmentation and mitophagy. Infection of epithelial cells with either wildtype EPEC or an isogenic espZ deletion mutant (ΔespZ) robustly upregulated FIS1 abundance, but a marked increase in mitochondrial fragmentation and mitophagy was seen only in ΔespZ-infected cells. FIS1-depleted cells were protected against ΔespZ-induced fission, and EspZ-expressing transfected epithelial cells were protected against pharmacologically induced mitochondrial fission and membrane potential disruption. Thus, EspZ interacts with FIS1 and blocks mitochondrial fragmentation and mitophagy. In contrast to WT EPEC, ΔespH-infected epithelial cells had minimal FIS1 upregulation and exhibited hyperfused mitochondria. Consistent with the contrasting impacts on organelle shape, mitochondrial membrane potential was preserved in ΔespH-infected cells, but profoundly disrupted in ΔespZ-infected cells. Collectively, our studies reveal hitherto unappreciated roles for two essential EPEC virulence factors in the temporal and dynamic regulation of host mitochondrial biology.


Bacterial pathogens strategically manipulate host cell structures and functions during the process of colonization and expansion, and this eventually contributes to disease symptoms. The diarrhea-causing pathogen enteropathogenic Escherichia coli (EPEC) secretes proteins into host cells to alter their behavior. Two secreted proteins, EspZ and EspH, were previously shown to be essential for causing disease in animal models. In this study, we demonstrate that interplay between EspZ/EspH and host factors modulates the structure and function of host cell mitochondria. Among their various roles, mitochondria generate energy, produce important biomolecules, and protect cells from damage. EPEC infection of epithelial cells results in increased abundance of a key mitochondrial outer-membrane protein, FIS1. FIS1 plays a housekeeping role by breaking down unhealthy mitochondria and targeting them for elimination from cells. In the early stages of infection, EspZ interacts with FIS1 and blocks its action, thereby protecting the host mitochondrial network and consequently, enhancing host cell viability. Our studies are consistent with a model wherein EspZ-dependent preservation of mitochondrial integrity early in infection allows for bacterial colonization. Later in infection, however, EspH-dependent increase in FIS1 results in significant mitochondrial fragmentation and host cell death; this likely facilitates pathogen dispersal. Taken together, EspZ and EspH dynamically impact host biology, and consequently, infection outcomes. Overall, an appreciation of the mechanisms by which EspZ and EspH manipulate host cells could eventually lead to host-directed interventions for EPEC diarrhea, which is currently not vaccine-preventable.


Subject(s)
Enteropathogenic Escherichia coli , Gastrointestinal Microbiome , Enteropathogenic Escherichia coli/genetics
2.
Sci Rep ; 10(1): 22135, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33335199

ABSTRACT

Clostridioides difficile infection (CDI) is a major healthcare-associated diarrheal disease. Consistent with trends across the United States, C. difficile RT106 was the second-most prevalent molecular type in our surveillance in Arizona from 2015 to 2018. A representative RT106 strain displayed robust virulence and 100% lethality in the hamster model of acute CDI. We identified a unique 46 KB genomic island (GI1) in all RT106 strains sequenced to date, including those in public databases. GI1 was not found in its entirety in any other C. difficile clade, or indeed, in any other microbial genome; however, smaller segments were detected in Enterococcus faecium strains. Molecular clock analyses suggested that GI1 was horizontally acquired and sequentially assembled over time. GI1 encodes homologs of VanZ and a SrtB-anchored collagen-binding adhesin, and correspondingly, all tested RT106 strains had increased teicoplanin resistance, and a majority displayed collagen-dependent biofilm formation. Two additional genomic islands (GI2 and GI3) were also present in a subset of RT106 strains. All three islands are predicted to encode mobile genetic elements as well as virulence factors. Emergent phenotypes associated with these genetic islands may have contributed to the relatively rapid expansion of RT106 in US healthcare and community settings.


Subject(s)
Clostridioides difficile/classification , Clostridioides difficile/genetics , Genome, Bacterial , Genomic Islands , Genomics , Phenotype , Phylogeny , Ribotyping , Animals , Anti-Bacterial Agents/pharmacology , Arizona/epidemiology , Clostridioides difficile/drug effects , Clostridioides difficile/isolation & purification , Clostridium Infections/epidemiology , Clostridium Infections/microbiology , Cricetinae , Cross Infection/epidemiology , Drug Resistance, Bacterial , Genetic Variation , Genomics/methods , Genotype , Humans , Microbial Sensitivity Tests , Prevalence , Public Health Surveillance , Ribotyping/methods
3.
mBio ; 10(1)2019 01 08.
Article in English | MEDLINE | ID: mdl-30622186

ABSTRACT

Clostridium difficile infection (CDI) is a major public health threat worldwide. The use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with enhanced susceptibility to and severity of CDI; however, the mechanisms driving this phenomenon have not been elucidated. NSAIDs alter prostaglandin (PG) metabolism by inhibiting cyclooxygenase (COX) enzymes. Here, we found that treatment with the NSAID indomethacin prior to infection altered the microbiota and dramatically increased mortality and the intestinal pathology associated with CDI in mice. We demonstrated that in C. difficile-infected animals, indomethacin treatment led to PG deregulation, an altered proinflammatory transcriptional and protein profile, and perturbed epithelial cell junctions. These effects were paralleled by increased recruitment of intestinal neutrophils and CD4+ cells and also by a perturbation of the gut microbiota. Together, these data implicate NSAIDs in the disruption of protective COX-mediated PG production during CDI, resulting in altered epithelial integrity and associated immune responses.IMPORTANCEClostridium difficile infection (CDI) is a spore-forming anaerobic bacterium and leading cause of antibiotic-associated colitis. Epidemiological data suggest that use of nonsteroidal anti-inflammatory drugs (NSAIDs) increases the risk for CDI in humans, a potentially important observation given the widespread use of NSAIDs. Prior studies in rodent models of CDI found that NSAID exposure following infection increases the severity of CDI, but mechanisms to explain this are lacking. Here we present new data from a mouse model of antibiotic-associated CDI suggesting that brief NSAID exposure prior to CDI increases the severity of the infectious colitis. These data shed new light on potential mechanisms linking NSAID use to worsened CDI, including drug-induced disturbances to the gut microbiome and colonic epithelial integrity. Studies were limited to a single NSAID (indomethacin), so future studies are needed to assess the generalizability of our findings and to establish a direct link to the human condition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Clostridium Infections/mortality , Clostridium Infections/pathology , Gastrointestinal Microbiome/drug effects , Indomethacin/adverse effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Indomethacin/administration & dosage , Mice , Neutrophils/immunology , Prostaglandins/analysis , Survival Analysis
4.
Gut Microbes ; 10(2): 241-245, 2019.
Article in English | MEDLINE | ID: mdl-30183504

ABSTRACT

Desmosomes are junctional protein complexes that confer strong adhesive capacity to adjacent host cells. In a recent study, we showed that enteropathogenic Escherichia coli (EPEC) disrupts desmosomes, weakens cell-cell adhesion and perturbs barrier function of intestinal epithelial (C2BBe) cells. Desmosomal damage was dependent on the EPEC effector protein EspH and its inhibitory effect on Rho GTPases. EspH-mediated Rho inactivation resulted in retraction of keratin intermediate filaments and degradation of desmosomal cadherins. Immunofluorescence studies of EPEC-infected C2BBe cells revealed keratin retraction towards the nucleus coincident with significant cytoplasmic redistribution of the desmosomal cadherin desmoglein-2 (DSG2). In this addendum, we expand on how EPEC-induced keratin retraction leads to loss of DSG2 anchoring at the junctions, and show that maturity of the epithelial cell monolayer impacts the fate of desmosomes during infection.


Subject(s)
Desmosomes/microbiology , Enteropathogenic Escherichia coli/physiology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiology , Cell Adhesion , Cell Line, Tumor , Desmoglein 2/metabolism , Desmosomes/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Host-Pathogen Interactions , Humans , Intestinal Mucosa/metabolism , Keratins/metabolism
5.
Front Microbiol ; 9: 2080, 2018.
Article in English | MEDLINE | ID: mdl-30233548

ABSTRACT

Morbidity and mortality attributed to Clostridium difficile infection (CDI) have increased over the past 20 years. Currently, antibiotics are the only US FDA-approved treatment for primary C. difficile infection, and these are, ironically, associated with disease relapse and the threat of burgeoning drug resistance. We previously showed that non-toxin virulence factors play key roles in CDI, and that colonization factors are critical for disease. Specifically, a C. difficile adhesin, Surface Layer Protein A (SlpA) is a major contributor to host cell attachment. In this work, we engineered Syn-LAB 2.0 and Syn-LAB 2.1, two synthetic biologic agents derived from lactic acid bacteria, to stably and constitutively express a host-cell binding fragment of the C. difficile adhesin SlpA on their cell-surface. Both agents harbor conditional suicide plasmids expressing a codon-optimized chimera of the lactic acid bacterium's cell-wall anchoring surface-protein domain, fused to the conserved, highly adherent, host-cell-binding domain of C. difficile SlpA. Both agents also incorporate engineered biocontrol, obviating the need for any antibiotic selection. Syn-LAB 2.0 and Syn-LAB 2.1 possess positive biophysical and in vivo properties compared with their parental antecedents in that they robustly and constitutively display the SlpA chimera on their cell surface, potentiate human intestinal epithelial barrier function in vitro, are safe, tolerable and palatable to Golden Syrian hamsters and neonatal piglets at high daily doses, and are detectable in animal feces within 24 h of dosing, confirming robust colonization. In combination, the engineered strains also delay (in fixed doses) or prevent (when continuously administered) death of infected hamsters upon challenge with high doses of virulent C. difficile. Finally, fixed-dose Syn-LAB ameliorates diarrhea in a non-lethal model of neonatal piglet enteritis. Taken together, our findings suggest that the two synthetic biologics may be effectively employed as non-antibiotic interventions for CDI.

6.
Cell Mol Gastroenterol Hepatol ; 6(2): 163-180, 2018.
Article in English | MEDLINE | ID: mdl-30003123

ABSTRACT

BACKGROUND & AIMS: The diarrheagenic pathogen, enteropathogenic Escherichia coli (EPEC), uses a type III secretion system to deliver effector molecules into intestinal epithelial cells (IECs). While exploring the basis for the lateral membrane separation of EPEC-infected IECs, we observed infection-induced loss of the desmosomal cadherin desmoglein-2 (DSG2). We sought to identify the molecule(s) involved in, and delineate the mechanisms and consequences of, EPEC-induced DSG2 loss. METHODS: DSG2 abundance and localization was monitored via immunoblotting and immunofluorescence, respectively. Junctional perturbations were visualized by electron microscopy, and cell-cell adhesion was assessed using dispase assays. EspH alanine-scan mutants as well as pharmacologic agents were used to evaluate impacts on desmosomal alterations. EPEC-mediated DSG2 loss, and its impact on bacterial colonization in vivo, was assessed using a murine model. RESULTS: The secreted virulence protein EspH mediates EPEC-induced DSG2 degradation, and contributes to desmosomal perturbation, loss of cell junction integrity, and barrier disruption in infected IECs. EspH sequesters Rho guanine nucleotide exchange factors and inhibits Rho guanosine triphosphatase signaling; EspH mutants impaired for Rho guanine nucleotide exchange factor interaction failed to inhibit RhoA or deplete DSG2. Cytotoxic necrotizing factor 1, which locks Rho guanosine triphosphatase in the active state, jasplakinolide, a molecule that promotes actin polymerization, and the lysosomal inhibitor bafilomycin A, respectively, rescued infected cells from EPEC-induced DSG2 loss. Wild-type EPEC, but not an espH-deficient strain, colonizes mouse intestines robustly, widens paracellular junctions, and induces DSG2 re-localization in vivo. CONCLUSIONS: Our studies define the mechanism and consequences of EPEC-induced desmosomal alterations in IECs. These perturbations contribute to the colonization and virulence of EPEC, and likely related pathogens.

7.
Compr Physiol ; 8(2): 823-842, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29687905

ABSTRACT

The passive and regulated movement of ions, solutes, and water via spaces between cells of the epithelial monolayer plays a critical role in the normal intestinal functioning. This paracellular pathway displays a high level of structural and functional specialization, with the membrane-spanning complexes of the tight junctions, adherens junctions, and desmosomes ensuring its integrity. Tight junction proteins, like occludin, tricellulin, and the claudin family isoforms, play prominent roles as barriers to unrestricted paracellular transport. The past decade has witnessed major advances in our understanding of the architecture and function of epithelial tight junctions. While it has been long appreciated that microbes, notably bacterial and viral pathogens, target and disrupt junctional complexes and alter paracellular permeability, the precise mechanisms remain to be defined. Notably, renewed efforts will be required to interpret the available data on pathogen-mediated barrier disruption in the context of the most recent findings on tight junction structure and function. While much of the focus has been on pathogen-induced dysregulation of junctional complexes, commensal microbiota and their products may influence paracellular permeability and contribute to the normal physiology of the gut. Finally, microbes and their products have become important tools in exploring host systems, including the junctional properties of epithelial cells. © 2018 American Physiological Society. Compr Physiol 8:823-842, 2018.


Subject(s)
Bacteria/pathogenicity , Gastrointestinal Microbiome/physiology , Tight Junctions/physiology , Animals , Bacterial Infections/physiopathology , Biological Transport/physiology , Humans , Intestinal Mucosa/metabolism , Permeability
8.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G374-80, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24904077

ABSTRACT

The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) dynamically modulates the survival of infected host intestinal epithelial cells. In the initial stages of infection, several prosurvival signaling events are activated in host cells. These include the phosphorylation of epidermal growth factor receptor (EGFR) and the consequent activation of the phosphatidylinositol-3 kinase/Akt pathway. While studying this pathway in infected epithelial cells, we observed EGFR depletion at later stages of infection, followed subsequently by a decrease in phospho-EGFR. EGFR loss was not dependent on receptor phosphorylation, or on canonical proteasome- and lysosome-dependent processes. Although a type III secretion mutant (ΔescN) stimulated EGFR phosphorylation, it failed to induce receptor degradation. To identify the specific EPEC effector molecule(s) that influenced EGFR stability, epithelial cells infected with isogenic mutant EPEC strains were examined. An EPEC ΔespF strain failed to induce EGFR degradation, whereas EPEC ΔespZ accentuated receptor loss in infected cells. Given the known and contrasting effects of EspF and EspZ on caspase activation, and the known role of proteases in cleaving EGFR, we explored the effect of caspase inhibitors on infection-dependent EGFR loss. The pan-caspase inhibitor Q-VD-OPh blocked EPEC-induced EGFR cleavage in a dose-dependent manner. Taken together, our data suggest that EPEC EspF stimulates caspase-dependent EGFR cleavage and loss, whereas EspZ inhibits this process. Whereas EGFR phosphorylation contributes to the survival of host cells early in infection, EspF-driven caspase activation and consequent EGFR loss likely induce a precipitous increase in host cell death later in the infectious process.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , ErbB Receptors/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Signal Transduction , Caco-2 Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase Inhibitors/pharmacology , Cell Death , Dose-Response Relationship, Drug , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Epithelial Cells/drug effects , Epithelial Cells/pathology , ErbB Receptors/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Host-Pathogen Interactions , Humans , Intestines/drug effects , Intestines/pathology , Intracellular Signaling Peptides and Proteins , Mutation , Phosphorylation , Protein Stability , Signal Transduction/drug effects , Time Factors
9.
Infect Immun ; 80(11): 3850-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22907816

ABSTRACT

The diarrheagenic pathogen enteropathogenic Escherichia coli (EPEC) limits the death of infected enterocytes early in infection. A number of bacterial molecules and host signaling pathways contribute to the enhanced survival of EPEC-infected host cells. EspZ, a type III secreted effector protein that is unique to EPEC and related "attaching and effacing" (A/E) pathogens, plays a role in limiting host cell death, but the precise host signaling pathways responsible for this phenotype are not known. We hypothesized that EspZ contributes to the survival of infected intestinal epithelial cells by interfering with apoptosis. Consistent with previous studies, scanning electron microscopy analysis of intestinal epithelial cells infected with an EPEC espZ mutant (ΔespZ) showed increased levels of apoptotic and necrotic cells compared to cells infected with the isogenic parent strain. Correspondingly, higher levels of cytosolic cytochrome c and increased activation of caspases 9, 7, and 3 were observed for ΔespZ strain-infected cells compared to wild-type (WT) EPEC-infected cells. Finally, espZ-transfected epithelial cells were significantly protected from staurosporine-induced, but not tumor necrosis factor alpha (TNF-α)/cycloheximide-induced, apoptosis. Thus, EspZ contributes to epithelial cell survival by mechanisms that include the inhibition of the intrinsic apoptotic pathway. The enhanced survival of infected enterocytes by molecules such as EspZ likely plays a key role in optimal colonization by A/E pathogens.


Subject(s)
Apoptosis/physiology , Enteropathogenic Escherichia coli/metabolism , Epithelial Cells/metabolism , Escherichia coli Proteins/metabolism , Intestinal Mucosa/metabolism , Cells, Cultured , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Humans , Intestines/microbiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...