Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Sci Rep ; 13(1): 7024, 2023 04 29.
Article in English | MEDLINE | ID: mdl-37120613

ABSTRACT

ANR (AraC negative regulators) are a novel class of small regulatory proteins commonly found in enteric pathogens. Aar (AggR-activated regulator), the best-characterized member of the ANR family, regulates the master transcriptional regulator of virulence AggR and the global regulator HNS in enteroaggregative Escherichia coli (EAEC) by protein-protein interactions. On the other hand, Rnr (RegA-negative regulator) is an ANR homolog identified in attaching and effacing (AE) pathogens, including Citrobacter rodentium and enteropathogenic Escherichia coli (EPEC), sharing only 25% identity with Aar. We previously found that C. rodentium lacking Rnr exhibits prolonged shedding and increased gut colonization in mice compared to the parental strain. To gain mechanistic insights into this phenomenon, we characterized the regulatory role of Rnr in the virulence of prototype EPEC strain E2348/69 by genetic, biochemical, and human organoid-based approaches. Accordingly, RNA-seq analysis revealed more than 500 genes differentially regulated by Rnr, including the type-3 secretion system (T3SS). The abundance of EspA and EspB in whole cells and bacterial supernatants confirmed the negative regulatory activity of Rnr on T3SS effectors. We found that besides HNS and Ler, twenty-six other transcriptional regulators were also under Rnr control. Most importantly, the deletion of aar in EAEC or rnr in EPEC increases the adherence of these pathogens to human intestinal organoids. In contrast, the overexpression of ANR drastically reduces bacterial adherence and the formation of AE lesions in the intestine. Our study suggests a conserved regulatory mechanism and a central role of ANR in modulating intestinal colonization by these enteropathogens despite the fact that EAEC and EPEC evolved with utterly different virulence programs.


Subject(s)
Enteropathogenic Escherichia coli , Escherichia coli Infections , Escherichia coli Proteins , Humans , Animals , Mice , Virulence/genetics , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Transcription Factors
2.
PLoS Pathog ; 16(8): e1008776, 2020 08.
Article in English | MEDLINE | ID: mdl-32845938

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is a diarrheagenic pathotype associated with traveler's diarrhea, foodborne outbreaks and sporadic diarrhea in industrialized and developing countries. Regulation of virulence in EAEC is mediated by AggR and its negative regulator Aar. Together, they control the expression of at least 210 genes. On the other hand, we observed that about one third of Aar-regulated genes are related to metabolism and transport. In this study we show the AggR/Aar duo controls the metabolism of lipids. Accordingly, we show that AatD, encoded in the AggR-regulated aat operon (aatPABCD) is an N-acyltransferase structurally similar to the essential Apolipoprotein N-acyltransferase Lnt and is required for the acylation of Aap (anti-aggregation protein). Deletion of aatD impairs post-translational modification of Aap and causes its accumulation in the bacterial periplasm. trans-complementation of 042aatD mutant with the AatD homolog of ETEC or with the N-acyltransferase Lnt reestablished translocation of Aap. Site-directed mutagenesis of the E207 residue in the putative acyltransferase catalytic triad disrupted the activity of AatD and caused accumulation of Aap in the periplasm due to reduced translocation of Aap at the bacterial surface. Furthermore, Mass spectroscopy revealed that Aap is acylated in a putative lipobox at the N-terminal of the mature protein, implying that Aap is a lipoprotein. Lastly, deletion of aatD impairs bacterial colonization of the streptomycin-treated mouse model. Our findings unveiled a novel N-acyltransferase family associated with bacterial virulence, and that is tightly regulated by AraC/XylS regulators in the order Enterobacterales.


Subject(s)
Acetyltransferases/metabolism , AraC Transcription Factor/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Escherichia coli/pathogenicity , Gene Expression Regulation, Bacterial , Acetyltransferases/genetics , Acylation , Animals , AraC Transcription Factor/chemistry , AraC Transcription Factor/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Male , Mice , Mice, Inbred BALB C , Operon , Phylogeny , Protein Conformation , Virulence
3.
Infect Immun ; 88(9)2020 08 19.
Article in English | MEDLINE | ID: mdl-32631917

ABSTRACT

Symptomatic and asymptomatic infection with the diarrheal pathogen enteroaggregative Escherichia coli (EAEC) is associated with growth faltering in children in developing settings. The mechanism of this association is unknown, emphasizing a need for better understanding of the interactions between EAEC and the human gastrointestinal mucosa. In this study, we investigated the role of the aggregative adherence fimbriae II (AAF/II) in EAEC adherence and pathogenesis using human colonoids and duodenal enteroids. We found that a null mutant in aafA, the major subunit of AAF/II, adhered significantly less than wild-type (WT) EAEC strain 042, and adherence was restored in a complemented strain. Immunofluorescence confocal microscopy of differentiated colonoids, which produce an intact mucus layer comprised of the secreted mucin MUC2, revealed bacteria at the epithelial surface and within the MUC2 layer. The WT strain adhered to the epithelial surface, whereas the aafA deletion strain remained within the MUC2 layer, suggesting that the presence or absence of AAF/II determines both the abundance and location of EAEC adherence. In order to determine the consequences of EAEC adherence on epithelial barrier integrity, colonoid monolayers were exposed to EAEC constructs expressing or lacking aafA Colonoids infected with WT EAEC had significantly decreased epithelial resistance, an effect that required AAF/II, suggesting that binding of EAEC to the epithelium is necessary to impair barrier function. In summary, we show that production of AAF/II is critical for adherence and barrier disruption in human colonoids, suggesting a role for this virulence factor in EAEC colonization of the gastrointestinal mucosa.


Subject(s)
Adhesins, Escherichia coli/immunology , Epithelial Cells/microbiology , Escherichia coli/immunology , Fimbriae, Bacterial/immunology , Host Microbial Interactions/immunology , Organoids/microbiology , Adhesins, Escherichia coli/genetics , Bacterial Adhesion , Colon/immunology , Colon/metabolism , Colon/microbiology , Colony Count, Microbial , Duodenum/immunology , Duodenum/metabolism , Duodenum/microbiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Fimbriae, Bacterial/genetics , Gene Deletion , Gene Expression Regulation , Genetic Complementation Test , Host Microbial Interactions/genetics , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mucin-2/genetics , Mucin-2/immunology , Organoids/immunology , Organoids/metabolism , Signal Transduction
4.
Sci Rep ; 10(1): 10533, 2020 06 29.
Article in English | MEDLINE | ID: mdl-32601325

ABSTRACT

EAEC is a common cause of diarrheal illness worldwide. Pathogenesis is believed to occur in the ileum and colon, where the bacteria adhere and form a robust aggregating biofilm. Among the multiple virulence factors produced by EAEC, the Pic serine protease has been implicated in bacterial colonization by virtue of its mucinolytic activity. Hence, a potential role of Pic in mucus barrier disruption during EAEC infection has been long postulated. In this study, we used human colonoids comprising goblet cells and a thick mucin barrier as an intestinal model to investigate Pic's roles during infection with EAEC. We demonstrated the ability of purified Pic, but not a protease defective Pic mutant to degrade MUC2. Western blot and confocal microscopy analysis revealed degradation of the MUC2 layer in colonoids infected with EAEC, but not with its isogenic EAECpic mutant. Wild-type and MUC2-knockdown colonoids infected with EAEC strains exposed a differential biofilm distribution, greater penetration of the mucus layer and increased colonization of the colonic epithelium by Wild-type EAEC than its isogenic Pic mutant. Higher secretion of pro-inflammatory cytokines was seen in colonoids infected with EAEC than EAECpic. Although commensal E. coli expressing Pic degraded MUC2, it did not show improved mucus layer penetration or colonization of the colonic epithelium. Our study demonstrates a role of Pic in MUC2 barrier disruption in the human intestine and shows that colonoids are a reliable system to study the interaction of pathogens with the mucus layer.


Subject(s)
Colon/microbiology , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli , Intestinal Mucosa/microbiology , Serine Endopeptidases/metabolism , Colon/metabolism , Goblet Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Mucins/metabolism
5.
PLoS Negl Trop Dis ; 14(5): e0008274, 2020 05.
Article in English | MEDLINE | ID: mdl-32357189

ABSTRACT

Multidrug-resistant Escherichia coli ST131 fimH30 responsible for extra-intestinal pathogenic (ExPEC) infections is globally distributed. However, the occurrence of a subclone fimH27 of ST131 harboring both ExPEC and enteroaggregative E. coli (EAEC) related genes and belonging to commonly reported O25:H4 and other serotypes causing bacteremia in African children remain unknown. We characterized 325 E. coli isolates causing bacteremia in Mozambican children between 2001 and 2014 by conventional multiplex polymerase chain reaction and whole genome sequencing. Incidence rate of EAEC bacteremia was calculated among cases from the demographic surveillance study area. Approximately 17.5% (57/325) of isolates were EAEC, yielding an incidence rate of 45.3 episodes/105 children-years-at-risk among infants; and 44 of isolates were sequenced. 72.7% (32/44) of sequenced strains contained simultaneously genes associated with ExPEC (iutA, fyuA and traT); 88.6% (39/44) harbored the aggregative adherence fimbriae type V variant (AAF/V). Sequence type ST-131 accounted for 84.1% (37/44), predominantly belonging to serotype O25:H4 (59% of the 37); 95.6% (35/44) harbored fimH27. Approximately 15% (6/41) of the children died, and five of the six yielded ST131 strains (83.3%) mostly (60%; 3/5) due to serotypes other than O25:H4. We report the emergence of a new subclone of ST-131 E. coli strains belonging to O25:H4 and other serotypes harboring both ExPEC and EAEC virulence genes, including agg5A, associated with poor outcome in bacteremic Mozambican children, suggesting the need for prompt recognition for appropriate management.


Subject(s)
Adhesins, Escherichia coli/genetics , Bacteremia/microbiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli/classification , Fimbriae, Bacterial/genetics , Genotype , Trans-Activators/genetics , Adolescent , Bacteremia/epidemiology , Child , Child, Preschool , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Female , Humans , Incidence , Infant , Infant, Newborn , Male , Mozambique/epidemiology , Polymerase Chain Reaction , Serogroup , Whole Genome Sequencing
6.
Front Microbiol ; 10: 1965, 2019.
Article in English | MEDLINE | ID: mdl-31543869

ABSTRACT

Bacterial pathogens utilize a myriad of mechanisms to invade mammalian hosts, damage tissue sites, and evade the immune system. One essential strategy of Gram-negative bacteria is the secretion of virulence factors through both inner and outer membranes to reach a potential target. Most secretion systems are harbored in mobile elements including transposons, plasmids, pathogenicity islands, and phages, and Escherichia coli is one of the more versatile bacteria adopting this genetic information by horizontal gene transfer. Additionally, E. coli is a bacterial species with members of the commensal intestinal microbiota and pathogens associated with numerous types of infections such as intestinal, urinary, and systemic in humans and other animals. T6SS cluster plasticity suggests evolutionarily divergent systems were acquired horizontally. T6SS is a secretion nanomachine that is extended through the bacterial double membrane; from this apparatus, substrates are conveyed straight from the cytoplasm of the bacterium into a target cell or to the extracellular space. This nanomachine consists of three main complexes: proteins in the inner membrane that are T4SS component-like, the baseplate complex, and the tail complex, which are formed by components evolutionarily related to contractile bacteriophage tails. Advances in the T6SS understanding include the functional and structural characterization of at least 13 subunits (so-called core components), which are thought to comprise the minimal apparatus. So far, the main role of T6SS is on bacterial competition by using it to kill neighboring non-immune bacteria for which antibacterial proteins are secreted directly into the periplasm of the bacterial target after cell-cell contact. Interestingly, a few T6SSs have been associated directly to pathogenesis, e.g., roles in biofilm formation and macrophage survival. Here, we focus on the advances on T6SS from the perspective of E. coli pathotypes with emphasis in the secretion apparatus architecture, the mechanisms of pathogenicity of effector proteins, and the events of lateral gene transfer that led to its spread.

7.
Article in English | MEDLINE | ID: mdl-31131263

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) infections are one of the most frequent causes of persistent diarrhea in children, immunocompromised patients and travelers worldwide. The most prominent colonization factors of EAEC are aggregative adherence fimbriae (AAF). EAEC prototypical strain 042 harbors the AAF/II fimbriae variant, which mediates adhesion to intestinal epithelial cells and participates in the induction of an inflammatory response against this pathogen. However, the mechanism and the cell receptors implicated in eliciting this response have not been fully characterized. Since previous reports have shown that TLR4 recognize fimbriae from different pathogens, we evaluated the role of this receptor in the response elicited against EAEC by intestinal cells. Using a mutual antagonist against TLR2 and TLR4 (OxPAPC), we observed that blocking of these receptors significantly reduces the secretion of the inflammatory marker IL-8 in response to EAEC and AAF/II fimbrial extract in HT-29 cells. Using a TLR4-specific antagonist (TAK-242), we observed that the secretion of this cytokine was significantly reduced in HT-29 cells infected with EAEC or incubated with AAF/II fimbrial extract. We evaluated the participation of AAF/II fimbriae in the TLR4-mediated secretion of 38 cytokines, chemokines, and growth factors involved in inflammation. A reduction in the secretion of IL-8, GRO, and IL-4 was observed. Our results suggest that TLR4 participates in the secretion of several inflammation biomarkers in response to AAF/II fimbriae.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli/metabolism , Fimbriae, Bacterial/metabolism , Toll-Like Receptor 4/metabolism , Cytokines/metabolism , Escherichia coli Infections/metabolism , HT29 Cells , Humans , Inflammation , Interleukin-4 , Interleukin-8 , Intestines , Toll-Like Receptor 2/metabolism
8.
Toxins (Basel) ; 10(9)2018 09 01.
Article in English | MEDLINE | ID: mdl-30200426

ABSTRACT

One of the characteristic manifestations of Shiga-toxin-producing Escherichia coli (E. coli) infection in humans, including EHEC and Enteroaggregative E. coli O104:H4, is watery diarrhea. However, neither Shiga toxin nor numerous components of the type-3 secretion system have been found to independently elicit fluid secretion. We used the adult stem-cell-derived human colonoid monolayers (HCM) to test whether EHEC-secreted extracellular serine protease P (EspP), a member of the serine protease family broadly expressed by diarrheagenic E. coli can act as an enterotoxin. We applied the Ussing chamber/voltage clamp technique to determine whether EspP stimulates electrogenic ion transport indicated by a change in short-circuit current (Isc). EspP stimulates Isc in HCM. The EspP-stimulated Isc does not require protease activity, is not cystic fibrosis transmembrane conductance regulator (CFTR)-mediated, but is partially Ca2+-dependent. EspP neutralization with a specific antibody reduces its potency in stimulating Isc. Serine Protease A, secreted by Enteroaggregative E. coli, also stimulates Isc in HCM, but this current is CFTR-dependent. In conclusion, EspP stimulates colonic CFTR-independent active ion transport and may be involved in the pathophysiology of EHEC diarrhea. Serine protease toxins from E. coli pathogens appear to serve as enterotoxins, potentially significantly contributing to watery diarrhea.


Subject(s)
Bacterial Toxins/toxicity , Colon/drug effects , Escherichia coli Proteins/toxicity , Ion Transport/drug effects , Organoids/drug effects , Serine Endopeptidases/toxicity , Colon/physiology , Enterohemorrhagic Escherichia coli , Humans , Organoids/physiology
9.
Gut Microbes ; 9(3): 264-278, 2018.
Article in English | MEDLINE | ID: mdl-29543544

ABSTRACT

Enteroaggregative E. coli (EAEC) is associated with food-borne outbreaks of diarrhea and growth faltering among children in developing countries. A Shiga toxin-producing EAEC strain of serotype O104:H4 strain caused one of the largest outbreaks of a food-borne infection in Europe in 2011. The outbreak was traced to contaminated fenugreek sprouts, yet the mechanisms whereby such persistent contamination of sprouts could have occurred are not clear. We found that under ambient conditions of temperature and in minimal media, pathogenic Shiga toxin-producing EAEC O104:H4 227-11 and non-Shiga toxin-producing 042 strains both produce high levels of exopolysaccharide structures (EPS) that are released to the external milieu. The exopolysaccharide was identified as colanic acid (CA). Unexpectedly, Shiga-toxin producing EAEC strain 227-11 produced 3-6-fold higher levels of CA than the 042 strain, suggesting differential regulation of the CA in the two strains. The presence of CA was accompanied by the formation of large biofilm structures on the surface of sprouts. The wcaF-wza chromosomal locus was required for the synthesis of CA in EAEC 042. Deletion in the glycosyltransferase wcaE gene abolished the production of CA in 042, and resulted in diminished adherence to sprouts when co-cultured at ambient temperature. In conclusion, this work suggests that copious production of CA may contribute to persistence of EAEC in the environment and suggests a potential explanation for the large Shiga toxin-producing EAEC outbreak in 2011.


Subject(s)
Biofilms/growth & development , Escherichia coli/physiology , Polysaccharides, Bacterial/chemistry , Polysaccharides/biosynthesis , Seedlings/microbiology , Bacterial Proteins/genetics , Bile Acids and Salts/pharmacology , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Food Microbiology , Gene Deletion , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial/genetics , Genetic Complementation Test , Genome, Bacterial , Humans , Polysaccharides/genetics , Polysaccharides, Bacterial/genetics , Polysaccharides, Bacterial/ultrastructure
10.
Bio Protoc ; 8(8): e2802, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-34286021

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is a recognized cause of acute diarrhea among both children and adults worldwide. EAEC strains are characterized by the presence of aggregative adherence fimbriae (AAF), which play a key role in pathogenesis by mediating attachment to the intestinal mucosa and by triggering host inflammatory responses. The aggregative adherence fimbria II (AAF/II) is the most important adherence factor of EAEC prototype strain 042 (EAEC042) to intestinal cells. Multiple receptors for AAF/II on epithelial cells have been identified including the transmembrane signaling mucin Muc1. This protocol describes a method to measure adherence of EAEC strains to HEK293 cells expressing the Muc1 glycoprotein.

11.
Vet Microbiol ; 211: 180-188, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29102116

ABSTRACT

We previously developed attenuated rabbit enteropathogenic E. coli (rEPEC) strains which are effective oral vaccines against their parent pathogens by deleting ler, a global regulator of virulence genes. To use these strains as orally administered vectors to deliver other antigens we incorporated the B subunit of shiga-like toxin 1(Stx1) into the passenger domain of the autotransporter EspP expressed on a plasmid. Native EspP enters the periplasm where its passenger domain is exported to the bacterial surface through an outer membrane channel formed by its translocator domain, then cleaved and secreted. Since antigen localization may determine immunogenicity, we engineered derivatives of EspP expressing Stx1B- passenger domain fusions: 1. in cytoplasm 2. in periplasm, 3. surface-attached or 4. secreted. To determine which construct was most immunogenic, rabbits were immunized with attenuated O103 E. coli strain (E22 Δler) alone or expressing Stx1B in each of the above four cellular locations. IgG responses to Stx1B, and toxin-neutralizing antibodies were measured. Animals were challenged with a virulent rabbit Enterohemorrhagic E. coli (EHEC) strain of a different serogroup (O15) than the vaccine strain expressing Stx1 (RDEC-H19) and their clinical course observed. IgG responses to Stx1B subunit were induced in all animals vaccinated with the strain secreting Stx1B, in some vaccinated with surface-expressed Stx1B, but in not animals immunized with periplasmic or cytoplasmic Stx1B. Robust protection was observed only in the group immunized with the vaccine secreting Stx1B. Taken together, our data suggest that secretion of Stx1B, or other antigens, via an autotransporter, may maximize the protective response to live attenuated oral vaccine strains.


Subject(s)
Enteropathogenic Escherichia coli/immunology , Escherichia coli Infections/veterinary , Escherichia coli Vaccines/immunology , Immunization/veterinary , Type V Secretion Systems/metabolism , Animals , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Male , Plasmids/genetics , Rabbits , Shiga Toxin/genetics , Shiga Toxin/metabolism , Type V Secretion Systems/genetics , Virulence
12.
PLoS Pathog ; 13(8): e1006545, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28806780

ABSTRACT

The AraC Negative Regulators (ANR) comprise a large family of virulence regulators distributed among diverse clinically important Gram-negative pathogens, including Vibrio spp., Salmonella spp., Shigella spp., Yersinia spp., Citrobacter spp., and pathogenic E. coli strains. We have previously reported broad effects of the ANR members on regulators of the AraC/XylS family. Here, we interrogate possible broader effects of the ANR members on the bacterial transcriptome. Our studies focused on Aar (AggR-activated regulator), an ANR family archetype in enteroaggregative E. coli (EAEC) isolate 042. Transcriptome analysis of EAEC strain 042, 042aar and 042aar(pAar) identified more than 200 genes that were differentially expressed (+/- 1.5 fold, p<0.05). Most of those genes are located on the bacterial chromosome (195 genes, 92.85%), and are associated with regulation, transport, metabolism, and pathogenesis, based on the predicted annotation; a considerable number of Aar-regulated genes encoded for hypothetical proteins (46 genes, 21.9%) and regulatory proteins (25, 11.9%). Notably, the transcriptional expression of three histone-like regulators, H-NS (orf1292), H-NS homolog (orf2834) and StpA, was down-regulated in the absence of aar and may explain some of the effects of Aar on gene expression. By employing a bacterial two-hybrid system, LacZ reporter assays, pull-down and electrophoretic mobility shift assay (EMSA) analysis, we demonstrated that Aar binds directly to H-NS and modulates H-NS-induced gene silencing. Importantly, Aar was highly expressed in the mouse intestinal tract and was found to be necessary for maximal H-NS expression. In conclusion, this work further extends our knowledge of genes under the control of Aar and its biological relevance in vivo.


Subject(s)
AraC Transcription Factor/metabolism , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Gene Expression Regulation, Bacterial/physiology , Virulence/physiology , Animals , Electrophoretic Mobility Shift Assay , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Histones/metabolism , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction
13.
mBio ; 8(3)2017 06 06.
Article in English | MEDLINE | ID: mdl-28588132

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) causes diarrhea and intestinal inflammation worldwide. EAEC strains are characterized by the presence of aggregative adherence fimbriae (AAF), which play a key role in pathogenesis by mediating attachment to the intestinal mucosa and by triggering host inflammatory responses. Here, we identify the epithelial transmembrane mucin MUC1 as an intestinal host cell receptor for EAEC, demonstrating that AAF-mediated interactions between EAEC and MUC1 facilitate enhanced bacterial adhesion. We further demonstrate that EAEC infection also causes elevated expression of MUC1 in inflamed human intestinal tissues. Moreover, we find that MUC1 facilitates AAF-dependent migration of neutrophils across the epithelium in response to EAEC infection. Thus, we show for the first time a proinflammatory role for MUC1 in the host response to an intestinal pathogen.IMPORTANCE EAEC is a clinically important intestinal pathogen that triggers intestinal inflammation and diarrheal illness via mechanisms that are not yet fully understood. Our findings provide new insight into how EAEC triggers host inflammation and underscores the pivotal role of AAFs-the principal adhesins of EAEC-in driving EAEC-associated disease. Most importantly, our findings add a new dimension to the signaling properties of the transmembrane mucin MUC1. Mostly studied for its role in various forms of cancer, MUC1 is widely regarded as playing an anti-inflammatory role in response to infection with bacterial pathogens in various tissues. However, the role of MUC1 during intestinal infections has not been previously explored, and our results describe the first report of MUC1 as a proinflammatory factor following intestinal infection.


Subject(s)
Bacterial Adhesion , Epithelial Cells/microbiology , Escherichia coli/physiology , Fimbriae, Bacterial/immunology , Mucin-1/metabolism , Neutrophil Infiltration , Cell Movement , Diarrhea/microbiology , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Fimbriae, Bacterial/physiology , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Inflammation , Intestines/immunology , Intestines/microbiology , Intestines/physiopathology , Mucin-1/genetics , Neutrophils/physiology , Signal Transduction/immunology
14.
Nutr Res ; 39: 34-42, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28385287

ABSTRACT

In developing communities, intestinal infection is associated with poor weight gain and linear-growth failure. Prior translational animal models have focused on weight gain investigations into key contributors to linear growth failure have been lacking. We hypothesized that murine intestinal infection with Citrobacter rodentium would induce linear-growth failure associated with systemic inflammation and suppressed serum levels of insulin-like growth factor-1 (IGF-1). We evaluated 4 groups of mice infected or sham-infected on day-of-life 28: uninfected-controls, wild-type C rodentium-infected, partially-attenuated C rodentium-infected (with deletion of 3 serine protease genes involved in colonization), and pair-fed (given the amount of daily food consumed by the wild-type C rodentium group). Relative to the uninfected group, mice infected with wild-type C rodentium exhibited temporal associations of lower food intake, weight loss, linear-growth failure, higher IL-6 and TNF-α and lower IGF-1. However, relative to the pair-fed group, the C rodentium-infected group only differed significantly by linear growth and systemic inflammatory cytokines. Between post-infection days 15-20, the infected group exhibited resolution of systemic inflammation. Between days 16-20, both wild-type C rodentium and pair-fed groups exhibited rapid linear-growth velocities exceeding the uninfected and mutant C rodentium groups; during this time levels of IGF-1 increased to match the uninfected group. We submit this as a model providing important opportunities to study mechanisms of catch-up growth related to intestinal inflammation. We conclude that in addition to known effects of weight loss, infection with C rodentium induces linear-growth failure potentially related to systemic inflammation and low levels of IGF-1, with catch-up of linear growth following resolution of inflammation.


Subject(s)
Citrobacter rodentium , Colitis/complications , Colon/microbiology , Energy Intake/physiology , Growth Disorders/etiology , Inflammation/etiology , Insulin-Like Growth Factor I/metabolism , Animals , Colitis/metabolism , Colitis/microbiology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Eating , Growth Disorders/metabolism , Growth Disorders/microbiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice, Inbred C57BL/anatomy & histology , Tumor Necrosis Factor-alpha/metabolism , Weight Gain , Weight Loss
15.
Article in English | MEDLINE | ID: mdl-28008386

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) infections are still one of the most important etiologic pathogens of diarrhea in children worldwide. EAEC pathogenesis comprises three stages: adherence and colonization, production of toxins, and diarrhea followed by inflammation. Previous studies have demonstrated that EAEC strains have the ability to bind to fibronectin (FN); however, the role this extracellular matrix protein plays in the inflammatory response induced by EAEC remains unknown. In this study, we postulated that FN-mediated adherence of EAEC strains to epithelial cells increases the expression of pro-inflammatory genes. To verify this hypothesis, we infected HEp-2 and HT-29 cells, in both the presence and absence of FN, with EAEC reference strain 042. We quantified IL-8 secretion and the relative expression of a set of genes regulated by the NF-κB pathway. Although FN increased EAEC adherence, no changes in IL-8 protein secretion or IL8 gene expression were observed. Similar observations were found in HEp-2 cells transfected with FN-siRNA and infected with EAEC. To evaluate the involvement of AAF/II fimbriae, we infected HEp-2 and HT-29 cells, in both the presence and absence of FN, with an EAEC 042aafA mutant strain transformed with a plasmid harboring the native aafA gene with a site-directed mutation in Lys72 residue (K72A and K72R strains). No changes in IL-8 secretion were observed. Finally, SEM immunogold assay of cells incubated with FN and infected with EAEC revealed that AAF fimbriae can bind to cells either directly or mediated by FN. Our data suggests that FN participates in AAF/II fimbriae-mediated adherence of EAEC to epithelial cells, but not in the inflammatory response of cells infected by this pathogen.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/microbiology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Fibronectins/immunology , Inflammation/immunology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Cell Line , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fibronectins/genetics , Fibronectins/pharmacology , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression , Humans , Inflammation/genetics , Interleukin-8/genetics , Interleukin-8/metabolism , Mutagenesis, Site-Directed , NF-kappa B/genetics , NF-kappa B/metabolism
16.
Mol Microbiol ; 101(2): 314-32, 2016 07.
Article in English | MEDLINE | ID: mdl-27038276

ABSTRACT

AraC Negative Regulators (ANR) suppress virulence genes by directly down-regulating AraC/XylS members in Gram-negative bacteria. In this study, we sought to investigate the distribution and molecular mechanisms of regulatory function for ANRs among different bacterial pathogens. We identified more than 200 ANRs distributed in diverse clinically important gram negative pathogens, including Vibrio spp., Salmonella spp., Shigella spp., Yersinia spp., Citrobacter spp., enterotoxigenic (ETEC) and enteroaggregative E. coli (EAEC), and members of the Pasteurellaceae. By employing a bacterial two hybrid system, pull down assays and surface plasmon resonance (SPR) analysis, we demonstrate that Aar (AggR-activated regulator), a prototype member of the ANR family in EAEC, binds with high affinity to the central linker domain of AraC-like member AggR. ANR-AggR binding disrupted AggR dimerization and prevented AggR-DNA binding. ANR homologs of Vibrio cholerae, Citrobacter rodentium, Salmonella enterica and ETEC were capable of complementing Aar activity by repressing aggR expression in EAEC strain 042. ANR homologs of ETEC and Vibrio cholerae bound to AggR as well as to other members of the AraC family, including Rns and ToxT. The predicted proteins of all ANR members exhibit three highly conserved predicted α-helices. Site-directed mutagenesis studies suggest that at least predicted α-helices 2 and 3 are required for Aar activity. In sum, our data strongly suggest that members of the novel ANR family act by directly binding to their cognate AraC partners.


Subject(s)
AraC Transcription Factor/genetics , Genes, araC/genetics , AraC Transcription Factor/metabolism , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , Genes, araC/physiology , Gram-Negative Bacteria/genetics , Mutagenesis, Site-Directed , Phylogeny , Structure-Activity Relationship , Trans-Activators/metabolism , Transcription Factors/metabolism , Virulence/genetics
17.
Nat Immunol ; 17(2): 150-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595890

ABSTRACT

Mucosal surfaces are exposed to environmental substances and represent a major portal of entry for microorganisms. The innate immune system is responsible for early defense against infections and it is believed that the interferons (IFNs) constitute the first line of defense against viruses. Here we identify an innate antiviral pathway that works at epithelial surfaces before the IFNs. The pathway is activated independently of known innate sensors of viral infections through a mechanism dependent on viral O-linked glycans, which induce CXCR3 chemokines and stimulate antiviral activity in a manner dependent on neutrophils. This study therefore identifies a previously unknown layer of antiviral defense that exerts its action on epithelial surfaces before the classical IFN response is operative.


Subject(s)
Immunity, Innate , Interferons/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Cell Line , Chemokine CXCL10/biosynthesis , Disease Models, Animal , Female , Gene Expression , Glycosylation , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 2, Human/immunology , Humans , Interferons/genetics , Ligands , Mice , Mice, Knockout , Mucous Membrane/virology , Neutrophils/immunology , Neutrophils/metabolism , Polysaccharides/immunology , Receptors, CXCR3/deficiency , Receptors, CXCR3/metabolism , Vagina/immunology , Vagina/metabolism , Vagina/virology , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Viral Load , Virus Diseases/virology
18.
Infect Immun ; 83(5): 1893-903, 2015 May.
Article in English | MEDLINE | ID: mdl-25712927

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC), a leading cause of acute diarrhea, colonizes the intestine by means of adhesins. However, 15 to 50% of clinical isolates are negative for known adhesins, making it difficult to identify antigens for broad-coverage vaccines. The ETEC strain 1766a, obtained from a child with watery diarrhea in Chile, harbors the colonization factor CS23 but is negative for other known adhesins. One clone, derived from an ETEC 1766a genomic library (clone G10), did not produce CS23 yet was capable of adhering to Caco-2 cells. The goal of this study was to identify the gene responsible for this capacity. Random transposon-based mutagenesis allowed the identification of a 4,110-bp gene that codes for a homologue of the temperature-sensitive hemagglutinin (Tsh) autotransporter described in avian E. coli strains (97% identity, 90% coverage) and that is called TleA (Tsh-like ETEC autotransporter) herein. An isogenic ETEC 1766a strain with a tleA mutation showed an adhesion level similar to that of the wild-type strain, suggesting that the gene does not direct attachment to Caco-2 cells. However, expression of tleA conferred the capacity for adherence to nonadherent E. coli HB101. This effect coincided with the detection of TleA on the surface of nonpermeabilized bacteria, while, conversely, ETEC 1766a seems to secrete most of the produced autotransporter to the medium. On the other hand, TleA was capable of degrading bovine submaxillary mucin and leukocyte surface glycoproteins CD45 and P-selectin glycoprotein ligand 1 (PSGL-1). These results suggest that TleA promotes colonization of the intestinal epithelium and that it may modulate the host immune response.


Subject(s)
Adhesins, Bacterial/genetics , Adhesins, Escherichia coli/genetics , Bacterial Adhesion , Enterotoxigenic Escherichia coli/genetics , Enterotoxigenic Escherichia coli/physiology , Epithelial Cells/microbiology , Escherichia coli Proteins/genetics , Adhesins, Bacterial/metabolism , Adhesins, Escherichia coli/metabolism , Animals , Caco-2 Cells , Child, Preschool , Chile , DNA Transposable Elements , Diarrhea/microbiology , Enterotoxigenic Escherichia coli/isolation & purification , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gene Deletion , Humans , Infant , Infant, Newborn , Mutagenesis, Insertional
19.
PLoS One ; 9(9): e107920, 2014.
Article in English | MEDLINE | ID: mdl-25251283

ABSTRACT

The serine protease autotransporter from Enterobacteriaceae (SPATE) family, which number more than 25 proteases with apparent diverse functions, have been phylogenetically divided into two distinct classes, designated 1 and 2. We recently demonstrated that Pic and Tsh, two members of the class-2 SPATE family produced by intestinal and extraintestinal pathogenic E. coli, were able to cleave a number of O-glycosylated proteins on neutrophils and lymphocytes resulting in impaired leukocyte functions. Here we show that most members of the class-2 SPATE family have lectin-like properties and exhibit differential protease activity reliant on glycoprotein type and cell lineage. Protease activity was seen in virtually all tested O-glycosylated proteins including CD34, CD55, CD164, TIM1, TIM3, TIM4 and C1-INH. We also show that although SPATE proteins bound and cleaved glycoproteins more efficiently on granulocytes and monocytes, they also targeted glycoproteins on B, T and natural killer lymphocytes. Finally, we found that the characteristic domain-2 of class-2 SPATEs is not required for glycoprotease activity, but single amino acid mutations in Pic domain-1 to those residues naturally occurring in domain-1 of SepA, were sufficient to hamper Pic glycoprotease activity. This study shows that most class-2 SPATEs have redundant activities and suggest that they may function as immunomodulators at several levels of the immune system.


Subject(s)
Enterobacteriaceae Infections/immunology , Enterobacteriaceae/enzymology , Enterobacteriaceae/physiology , Host-Pathogen Interactions , Leukocytes/microbiology , Serine Proteases/immunology , Amino Acid Sequence , Cell Line , Cells, Cultured , Enterobacteriaceae/genetics , Enterobacteriaceae/immunology , Enterobacteriaceae Infections/metabolism , Glycoproteins/analysis , Glycoproteins/immunology , Glycoproteins/metabolism , Humans , Leukocytes/immunology , Leukocytes/metabolism , Models, Molecular , Molecular Sequence Data , Phylogeny , Proteolysis , Sequence Alignment , Serine Proteases/analysis , Serine Proteases/genetics , Serine Proteases/metabolism
20.
Biomed Res Int ; 2014: 781246, 2014.
Article in English | MEDLINE | ID: mdl-25177698

ABSTRACT

Adherence to the intestinal epithelia is a key feature in enteroaggregative Escherichia coli (EAEC) infection. The aggregative adherence fimbriae (AAFs) are involved in EAEC interaction with receptors at the surface of intestinal cells. We and others have demonstrated that fibronectin is a receptor for AAF/II fimbriae. Considering that the major cellular receptor of fibronectin is integrin α5ß1, in this study we evaluated the participation of this receptor in the fibronectin-mediated adherence of EAEC strain 042 to intestinal cells. We found that EAEC strain 042 has the ability to bind directly and indirectly to integrin α5ß1; direct binding was not mediated by AAF/II fimbriae and indirect binding was mediated by AAF/II and fibronectin. Coimmunoprecipitation assays confirmed the formation of the complex AafA/fibronectin/integrin α5ß1. To evaluate EAEC adherence to intestinal cells, T84 cells were incubated with fibronectin and an antibody that blocks the interaction region of integrin α5ß1 to fibronectin, the RGD site. Under these conditions, we found the number of adherent bacteria to epithelial cells significantly reduced. Additionally, fibronectin-mediated adherence of EAEC strain 042 was abolished in HEp-2 cells transfected with integrin α5 shRNA. Altogether, our data support the involvement of integrin α5ß1 in the fibronectin-mediated EAEC binding to intestinal cells.


Subject(s)
Bacterial Adhesion/physiology , Enterocytes/physiology , Escherichia coli/physiology , Fibronectins/metabolism , Integrin alpha5beta1/metabolism , Cell Line , Enterocytes/cytology , Escherichia coli/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL