Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 326(1): G25-G37, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37933481

ABSTRACT

Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in premature infants. Evidence indicates that bile acid homeostasis is disrupted during NEC: ileal bile acid levels are elevated in animals with experimental NEC, as is expression of the apical sodium-dependent bile acid transporter (Asbt). In addition, bile acids, which are synthesized in the liver, are extensively modified by the gut microbiome, including via the conversion of primary bile acids to more cytotoxic secondary forms. We hypothesized that the addition of bile acid-modifying bacteria would increase susceptibility to NEC in a neonatal rat model of the disease. The secondary bile acid-producing species Clostridium scindens exacerbated both incidence and severity of NEC. C. scindens upregulated the bile acid transporter Asbt and increased levels of intraenterocyte bile acids. Treatment with C. scindens also altered bile acid profiles and increased hydrophobicity of the ileal intracellular bile acid pool. The ability of C. scindens to enhance NEC requires bile acids, as pharmacological sequestration of ileal bile acids protects animals from developing disease. These findings indicate that bile acid-modifying bacteria can contribute to NEC pathology and provide additional evidence for the role of bile acids in the pathophysiology of experimental NEC.NEW & NOTEWORTHY Necrotizing enterocolitis (NEC), a life-threatening gastrointestinal emergency in premature infants, is characterized by dysregulation of bile acid homeostasis. We demonstrate that administering the secondary bile acid-producing bacterium Clostridium scindens enhances NEC in a neonatal rat model of the disease. C. scindens-enhanced NEC is dependent on bile acids and driven by upregulation of the ileal bile acid transporter Asbt. This is the first report of bile acid-modifying bacteria exacerbating experimental NEC pathology.


Subject(s)
Clostridiales , Enterocolitis, Necrotizing , Animals , Humans , Infant, Newborn , Rats , Bile Acids and Salts/metabolism , Enterocolitis, Necrotizing/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Up-Regulation , Disease Progression
2.
Front Med (Lausanne) ; 10: 1238159, 2023.
Article in English | MEDLINE | ID: mdl-37928470

ABSTRACT

Background: Clostridioides difficile Infection (CDI) is a healthcare-associated diarrheal disease prevalent worldwide. A common diagnostic algorithm relies on a two-step protocol that employs stool enzyme immunoassays (EIAs) to detect the pathogen, and its toxins, respectively. Active CDI is deemed less likely when the Toxin EIA result is negative, even if the pathogen-specific EIA is positive for C. difficile. We recently reported, however, that low-toxin-producing C. difficile strains recovered from Toxin-negative ('discrepant') clinical stool specimens can be fully pathogenic, and cause lethality in a rodent CDI model. To document frequency of discrepant CDI specimens, and evaluate C. difficile strain diversity, we performed longitudinal surveillance at a Southern Arizona tertiary-care hospital. Methods: Diarrheic stool specimens from patients with clinical suspicion of CDI were obtained over an eight-year period (2015-2022) from all inpatient and outpatient Units of a > 600-bed Medical Center in Southern Arizona. Clinical laboratory EIA testing identified C. difficile-containing specimens, and classified them as Toxin-positive or Toxin-negative. C. difficile isolates recovered from the stool specimens were DNA fingerprinted using an international phylogenetic lineage assignment system ("ribotyping"). For select isolates, toxin abundance in stationary phase supernatants of pure cultures was quantified via EIA. Results: Of 8,910 diarrheic specimens that underwent diagnostic testing, 1733 (19.4%) harbored C. difficile. Our major findings were that: (1) C. difficile prevalence and phylogenetic diversity was stable over the 8-year period; (2) toxigenic C. difficile was recovered from 69% of clinically Tox-neg ('discrepant') specimens; (3) the six most prevalent USA ribotypes were recovered in significant proportions (>60%) from Tox-neg specimens; and (4) toxin-producing C. difficile recovered from discrepant specimens produced less toxin than strains of the same ribotype isolated from non-discrepant specimens. Conclusion: Our study highlights the dominance of Toxin EIA-negative CDI specimens in a clinical setting and the high frequency of known virulent ribotypes in these specimens. Therefore, a careful reevaluation of the clinical relevance of diagnostically-discrepant specimens particularly in the context of missed CDI diagnoses and C. difficile persistence, is warranted.

3.
Int Rev Cell Mol Biol ; 377: 65-86, 2023.
Article in English | MEDLINE | ID: mdl-37268351

ABSTRACT

Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are human enteric pathogens that contribute significantly to morbidity and mortality worldwide. These extracellular pathogens attach intimately to intestinal epithelial cells and cause signature lesions by effacing the brush border microvilli, a property they share with other "attaching and effacing" (A/E) bacteria, including the murine pathogen Citrobacter rodentium. A/E pathogens use a specialized apparatus called a type III secretion system (T3SS) to deliver specific proteins directly into the host cytosol and modify host cell behavior. The T3SS is essential for colonization and pathogenesis, and mutants lacking this apparatus fail to cause disease. Thus, deciphering effector-induced host cell modifications is critical for understanding A/E bacterial pathogenesis. Several of the ∼20-45 effector proteins delivered into the host cell modify disparate mitochondrial properties, some via direct interactions with the mitochondria and/or mitochondrial proteins. In vitro studies have uncovered the mechanistic basis for the actions of some of these effectors, including their mitochondrial targeting, interaction partners, and consequent impacts on mitochondrial morphology, oxidative phosphorylation and ROS production, disruption of membrane potential, and intrinsic apoptosis. In vivo studies, mostly relying on the C. rodentium/mouse model, have been used to validate a subset of the in vitro observations; additionally, animal studies reveal broad changes to intestinal physiology that are likely accompanied by mitochondrial alterations, but the mechanistic underpinnings remain undefined. This chapter provides an overview of A/E pathogen-induced host alterations and pathogenesis, specifically focusing on mitochondria-targeted effects.


Subject(s)
Epithelial Cells , Mitochondria , Animals , Humans , Mice , Citrobacter rodentium/physiology
4.
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
5.
Emerg Microbes Infect ; 11(1): 1982-1993, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35880487

ABSTRACT

Clostridioides difficile is a leading cause of healthcare-associated infections worldwide. Currently, there is a lack of consensus for an optimal diagnostic method for C. difficile infection (CDI). Multi-step diagnostic algorithms use enzyme immunosorbent analysis (EIA)-based detection of C. difficile toxins TcdA/TcdB in stool, premised on the rationale that EIA toxin-negative (Tox-) patients have less severe disease and shorter diarrhoea duration. The aim of this study was to characterize toxigenic (i.e. tcdA/tcdB-positive) C. difficile strains isolated from diarrheic patient stool with an EIA Tox- (i.e. "discrepant") CDI diagnostic test result. Recovered strains were DNA fingerprinted (ribotyped), subjected to multiple toxin, genome and proteome evaluations, and assessed for virulence. Overall, of 1243 C. difficile-positive patient stool specimens from Southern Arizona hospitals, 31% were discrepant. For RT027 (the most prevalent ribotype)-containing specimens, 34% were discrepant; the corresponding RT027 isolates were cytotoxic to cultured fibroblasts, but their total toxin levels were comparable to, or lower than, the historic low-toxin-producing C. difficile strain CD630. Nevertheless, these low-toxin RT027 strains (LT-027) exhibited similar lethality to a clade-matched high-toxin RT027 strain in Golden Syrian hamsters, and heightened colonization and persistence in mice. Genomics and proteomics analyses of LT-027 strains identified unique genes and altered protein abundances, respectively, relative to high-toxin RT027 strains. Collectively, our data highlight the robust virulence of LT-027 C. difficile, provide a strong argument for reconsidering the clinical significance of a Tox- EIA result, and underscore the potential limitations of current diagnostic protocols.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Animals , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Clostridioides , Clostridioides difficile/genetics , Mice , Virulence
6.
Front Microbiol ; 13: 871152, 2022.
Article in English | MEDLINE | ID: mdl-35633701

ABSTRACT

The alternative sigma factor SigL (Sigma-54) facilitates bacterial adaptation to the extracellular environment by modulating the expression of defined gene subsets. A homolog of the gene encoding SigL is conserved in the diarrheagenic pathogen Clostridioides difficile. To explore the contribution of SigL to C. difficile biology, we generated sigL-disruption mutants (sigL::erm) in strains belonging to two phylogenetically distinct lineages-the human-relevant Ribotype 027 (strain BI-1) and the veterinary-relevant Ribotype 078 (strain CDC1). Comparative proteomics analyses of mutants and isogenic parental strains revealed lineage-specific SigL regulons. Concomitantly, loss of SigL resulted in pleiotropic and distinct phenotypic alterations in the two strains. Sporulation kinetics, biofilm formation, and cell surface-associated phenotypes were altered in CDC1 sigL::erm relative to the isogenic parent strain but remained unchanged in BI-1 sigL::erm. In contrast, secreted toxin levels were significantly elevated only in the BI-1 sigL::erm mutant relative to its isogenic parent. We also engineered SigL overexpressing strains and observed enhanced biofilm formation in the CDC1 background, and reduced spore titers as well as dampened sporulation kinetics in both strains. Thus, we contend that SigL is a key, pleiotropic regulator that dynamically influences C. difficile's virulence factor landscape, and thereby, its interactions with host tissues and co-resident microbes.

7.
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
8.
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
9.
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
10.
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.

11.
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.

12.
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
14.
PLoS Pathog ; 12(10): e1005946, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27741317

ABSTRACT

Clostridium difficile is a diarrheagenic pathogen associated with significant mortality and morbidity. While its glucosylating toxins are primary virulence determinants, there is increasing appreciation of important roles for non-toxin factors in C. difficile pathogenesis. Cell wall glycopolymers (CWGs) influence the virulence of various pathogens. Five C. difficile CWGs, including PSII, have been structurally characterized, but their biosynthesis and significance in C. difficile infection is unknown. We explored the contribution of a conserved CWG locus to C. difficile cell-surface integrity and virulence. Attempts at disrupting multiple genes in the locus, including one encoding a predicted CWG exporter mviN, were unsuccessful, suggesting essentiality of the respective gene products. However, antisense RNA-mediated mviN downregulation resulted in slight morphology defects, retarded growth, and decreased surface PSII deposition. Two other genes, lcpA and lcpB, with putative roles in CWG anchoring, could be disrupted by insertional inactivation. lcpA- and lcpB- mutants had distinct phenotypes, implying non-redundant roles for the respective proteins. The lcpB- mutant was defective in surface PSII deposition and shedding, and exhibited a remodeled cell surface characterized by elongated and helical morphology, aberrantly-localized cell septae, and an altered surface-anchored protein profile. Both lcpA- and lcpB- strains also displayed heightened virulence in a hamster model of C. difficile disease. We propose that gene products of the C. difficile CWG locus are essential, that they direct the production/assembly of key antigenic surface polysaccharides, and thereby have complex roles in virulence.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/ultrastructure , Clostridioides difficile/pathogenicity , Clostridioides difficile/ultrastructure , Clostridium Infections/virology , Virulence Factors/metabolism , Animals , Cell Wall/chemistry , Cricetinae , Disease Models, Animal , Fluorescent Antibody Technique , Immunoblotting , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mesocricetus , Microscopy, Electron , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Polysaccharides/chemistry , Polysaccharides/metabolism , Virulence
15.
Infect Immun ; 83(3): 1139-49, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25561713

ABSTRACT

Attaching and effacing (A/E) pathogens adhere intimately to intestinal enterocytes and efface brush border microvilli. A key virulence strategy of A/E pathogens is the type III secretion system (T3SS)-mediated delivery of effector proteins into host cells. The secreted protein EspZ is postulated to promote enterocyte survival by regulating the T3SS and/or by modulating epithelial signaling pathways. To explore the role of EspZ in A/E pathogen virulence, we generated an isogenic espZ deletion strain (ΔespZ) and corresponding cis-complemented derivatives of rabbit enteropathogenic Escherichia coli and compared their abilities to regulate the T3SS and influence host cell survival in vitro. For virulence studies, rabbits infected with these strains were monitored for bacterial colonization, clinical signs, and intestinal tissue alterations. Consistent with data from previous reports, espZ-transfected epithelial cells were refractory to infection-dependent effector translocation. Also, the ΔespZ strain induced greater host cell death than did the parent and complemented strains. In rabbit infections, fecal ΔespZ strain levels were 10-fold lower than those of the parent strain at 1 day postinfection, while the complemented strain was recovered at intermediate levels. In contrast to the parent and complemented mutants, ΔespZ mutant fecal carriage progressively decreased on subsequent days. ΔespZ mutant-infected animals gained weight steadily over the infection period, failed to show characteristic disease symptoms, and displayed minimal infection-induced histological alterations. Terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL) staining of intestinal sections revealed increased epithelial cell apoptosis on day 1 after infection with the ΔespZ strain compared to animals infected with the parent or complemented strains. Thus, EspZ-dependent host cell cytoprotection likely prevents epithelial cell death and sloughing and thereby promotes bacterial colonization.


Subject(s)
Enterocytes/microbiology , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Microvilli/microbiology , Animals , Apoptosis , Bacterial Load , Bacterial Secretion Systems/genetics , Enterocytes/pathology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/pathology , Escherichia coli Proteins/metabolism , Feces/microbiology , Gene Deletion , Gene Expression , Genetic Complementation Test , Host-Pathogen Interactions , Humans , Male , Microvilli/pathology , Rabbits , Virulence
16.
Gut Microbes ; 5(4): 439-40, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25222378

ABSTRACT

After dominating the earth in numbers and biomass for a good 3.5 billion years, microbes are now finally in the public consciousness. The vigorous surge in the study of microbes promises to yield novel approaches to treating diseases and maintaining health. In the middle of the deluge of scientific publications and lay articles, we pause to reflect on this trend.


Subject(s)
Health , Microbiology/trends , Microbiota/physiology , Biotechnology/methods , Biotechnology/trends , Drug Industry/methods , Drug Industry/trends , Humans
17.
Gut Microbes ; 5(3): 275-6, 2014.
Article in English | MEDLINE | ID: mdl-25068259

ABSTRACT

The robust expression of microbial pattern recognition receptors such as TLR4 and Nod2 in intestinal stem cells reflects an active communication dynamic between the host and the gut microbiota. A new study reveals that muramyl dipeptide, the bacterial cell wall peptidoglycan motif, activates Nod2 within crypt base columnar Lgr5-positive stem cells and promotes their survival. Apart from the immediate relevance to the growth of organoids for in vitro experiments, the study raises new questions about the molecular mechanisms whereby gut microbes influence intestinal physiology.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Bacteria/immunology , Cell Wall/immunology , Nod2 Signaling Adaptor Protein/metabolism , Peptidoglycan/immunology , Stem Cells/drug effects , Stem Cells/physiology , Bacteria/cytology , Cell Survival/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Stem Cells/enzymology
18.
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
19.
Gut Microbes ; 5(1): 3-4, 2014.
Article in English | MEDLINE | ID: mdl-24637595

ABSTRACT

Antibiotics and antibiotic resistance made news on several fronts in the past year. Many public health organizations, including the CDC, used terms such as "crisis", "catastrophic consequences", and "nightmare scenario" to highlight the rapid emergence and spread of antibiotic resistance. A report from the Pew Commission on Industrial Farm Animal Production, on the fifth anniversary of the publication of its landmark 2008 report, noted that state and federal legislative efforts to limit non-therapeutic use of antibiotics in animal production were thwarted by drug and food animal industries. In its lobbying disclosures, the Farm Bureau stated that such efforts to limit use of animal antibiotics were "based on emotion and no credible peer reviewed science." Meanwhile, there have been inexorable advances in our understanding of the molecular mechanisms by which antibiotics induce diversity and resistance in bacteria. This article reviews one study that probed the role of the bacterial general stress response in sub-inhibitory antibiotic-induced mutagenesis and antibiotic resistance.


Subject(s)
Animal Diseases/drug therapy , Anti-Bacterial Agents/therapeutic use , Bacteria/drug effects , Animal Diseases/microbiology , Animals , Bacteria/genetics , Bacteria/metabolism , Drug Resistance, Bacterial , Humans , Livestock/growth & development
20.
Gut Microbes ; 5(2): 143-5, 2014.
Article in English | MEDLINE | ID: mdl-24637797

ABSTRACT

Studies on bacterial virulence and host-pathogen interactions usually rely on type strains isolated from human or veterinary populations. For instance, the El Tor strain N16961 has been extensively used to characterize Vibrio cholerae virulence, while E2348/69 is a default choice for studies on enteropathogenic E. coli interactions with host cells. Subsequent to isolation, such strains are passaged under laboratory conditions in rich medium, and often genetically manipulated to induce specific mutations or other alterations. While the cumulative knowledge gained by focused studies on a limited number of bacterial isolates allows for rapid scientific progress, strain diversity resulting from prolonged propagation and manipulation in different laboratories may lead to a distorted perspective and, at times, confound attempts to replicate specific experiments. Exploring such aberrations is an inevitable and necessary, if not always welcome, part of scientific progress.


Subject(s)
Virulence/genetics , Escherichia coli/genetics , Escherichia coli/pathogenicity , Genetic Variation/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Laboratories , Reproducibility of Results , Vibrio cholerae/genetics , Vibrio cholerae/pathogenicity , Virulence/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...