Buscar en
Brazilian Journal of Microbiology
Toda la web
Inicio Brazilian Journal of Microbiology Diarrheagenic Escherichia coli
Journal Information
Vol. 47. Issue S1.
Pages 3-30 (December 2016)
Share
Share
Download PDF
More article options
Visits
6256
Vol. 47. Issue S1.
Pages 3-30 (December 2016)
Medical Microbiology
Open Access
Diarrheagenic Escherichia coli
Visits
6256
Tânia A.T. Gomesa,
Corresponding author
tatg.amaral@unifesp.br

Corresponding author.
, Waldir P. Eliasb, Isabel C.A. Scaletskya, Beatriz E.C. Gutha, Juliana F. Rodriguesc, Roxane M.F. Piazzab, Luís C.S. Ferreirac, Marina B. Martinezd
a Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Microbiologia, Imunologia e Parasitologia, São Paulo, SP, Brazil
b Instituto Butantan, Laboratório de Bacterologia, São Paulo, SP, Brazil
c Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Microbiologia, São Paulo, SP, Brazil
d Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, São Paulo, SP, Brazil
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Abstract

Most Escherichia coli strains live harmlessly in the intestines and rarely cause disease in healthy individuals. Nonetheless, a number of pathogenic strains can cause diarrhea or extraintestinal diseases both in healthy and immunocompromised individuals. Diarrheal illnesses are a severe public health problem and a major cause of morbidity and mortality in infants and young children, especially in developing countries. E. coli strains that cause diarrhea have evolved by acquiring, through horizontal gene transfer, a particular set of characteristics that have successfully persisted in the host. According to the group of virulence determinants acquired, specific combinations were formed determining the currently known E. coli pathotypes, which are collectively known as diarrheagenic E. coli. In this review, we have gathered information on current definitions, serotypes, lineages, virulence mechanisms, epidemiology, and diagnosis of the major diarrheagenic E. coli pathotypes.

Keywords:
Escherichia coli
Diarrhea
Pathogenic mechanisms
Virulence factor
Epidemiology
Full Text

The genus Escherichia, which was named after the German pediatrician Theodor Escherich, consists of facultative anaerobic Gram-negative bacilli that belong to the family Enterobacteriaceae.1 The genus type species Escherichia coli is widely distributed, where it is the major facultative anaerobe inhabiting the large intestine of humans and warm-blooded animals.2 Although most E. coli strains live harmlessly in the colon and seldom cause disease in healthy individuals, a number of pathogenic strains can cause intestinal and extraintestinal diseases both in healthy and immunocompromised individuals.3

Diarrheal illnesses are a severe public health problem and a major cause of morbidity and mortality in infants and young children.4 Low- and middle-income countries in Africa, Asia and Latin America are the most affected regions with diarrheal diseases occurring more often with lethal outcomes mainly due to poor living conditions (inadequate water supplies, poor environmental hygiene and sanitation, and insufficient education).5

E. coli strains involved in diarrheal diseases are one of the most important of the various etiological agents of diarrhea, where strains have evolved by the acquisition, through horizontal gene transfer, of a particular set of characteristics that have successfully persisted in the host.3,5,6 According to the group of virulence determinants acquired, specific combinations were formed determining the currently known E. coli pathotypes, which are collectively known as diarrheagenic E. coli (DEC).6 The DEC pathotypes differ regarding their preferential host colonization sites, virulence mechanisms, and the ensuing clinical symptoms and consequences, and are classified as enteropathogenic E. coli (EPEC), enterohemorrhagic (Shiga toxin-producing) E. coli (EHEC/STEC), enteroaggregative E. coli (EAEC), enterotoxigenic E. coli (ETEC), and enteroinvasive E. coli (EIEC).

Each of these pathotypes represents a group of clones that share specific virulence factors. Nevertheless, it should be pointed out that the plasticity of the E. coli genome has hindered the identification of certain E. coli isolates as a pathotype, because some isolates combine the main virulence characteristics of different pathotypes and are thus considered potentially more virulent hybrid pathogenic strains.5

Another less well-defined pathotype has been described, that is, the diffusely-adherent E. coli (DAEC) pathotype, which comprises strains that adhere to epithelial cells in a diffused distribution.6 Despite their classification as a group distinct from the other pathotypes, the designation of DAEC as a different DEC pathotype requires further epidemiological studies, which have been hampered by the difficulties in its identification and classification.5 Furthermore, certain E. coli strains that have been classified as the adherent invasive E. coli (AIEC) pathotype, comprise one of the potential agents for Crohn's disease (CD). CD is an inflammatory bowel disease (IBD), which is thought to be caused by a combination of factors (genetics, the intestinal microbiota, environmental factors, and enteric pathogens).7,8

Diarrheal episodes due to DEC infections are an important public health issue among children and adults in developing countries, because of their association with morbidity and mortality of children less than five years of age. It was our aim with this review to gather information on current definitions, serotypes, lineages, virulence mechanisms, epidemiology, and diagnosis of the major DEC pathotypes with emphasis on the studies conducted in Brazil.

Typical and atypical enteropathogenic E. coli

The term enteropathogenic E. coli (EPEC) was first used in 1995 by Neter et al.,9 to describe a number of E. coli strains epidemiologically related to a series of outbreaks of infantile diarrhea in the 1940s and 1950s.10,11 Originally identified by serotype, EPEC are now defined as those E. coli strains having the ability to cause diarrhea, to produce a histopathology on the intestinal epithelium known as the attaching and effacing (AE) lesion, and the inability to produce Shiga toxins and heat-labile (LT) or heat-stable (ST) enterotoxins.6

Improvements in techniques allowing a better understanding of the genome and virulence mechanisms among EPEC strains over the years have led to the sub-classification of EPEC into typical EPEC (tEPEC) and atypical EPEC (aEPEC).3,12 Typical EPEC strains causing human infectious diarrhea possess a large virulence plasmid known as the EPEC adherence factor (EAF) plasmid (pEAF), which encodes the type IV fimbriae called the bundle-forming pilus (BFP), while aEPEC do not possess this plasmid.6,12

The majority of tEPEC strains fall into well-recognized O serotypes. Classical EPEC O serogroups include O55, O86, O111, O114, O119, O127, and O142. The most common H antigens associated with EPEC are the H6 and H2 antigens.12–15 A less common EPEC type is H34, and a number of tEPEC strains are classified as non-motile (H-) in conventional tests. Typical EPEC strains belonging to non-classical serotypes have also been reported.12,16

Based on multilocus enzyme electrophoresis analysis (MLEE) of allelic differences between housekeeping genes, tEPEC strains have been subtyped into two major lineages, previously designated EPEC1 and EPEC2.13,14 EPEC1 includes widespread serotypes such as O55:H6 and O119:H6, whereas EPEC2 consists of serotypes with more limited occurrence such as O111:H2 and O114:H2. Based on a whole-genome phylogeny and analysis of type III secretion system (T3SS) effectors, tEPEC strains have been demonstrated to cluster in three main lineages, designated EPEC1, EPEC2, and EPEC4, which probably acquired the locus of enterocyte effacement (LEE) region and pEAF independently.17

In turn, aEPEC belong to a large diversity of classical and non-classical serotypes.12,16,18 Over 20% of strains of non-classical EPEC serotypes are O non-typeable and the O-typeable strains belong to more than 4200 different serotypes, with many non-motile and H non-typeable strains.12,18 Interestingly, it has been found that 35% of the aEPEC strains also belong to the tEPEC lineages.17 Thus, it has been hypothesized that at least some aEPEC may have originated from tEPEC strains that lost pEAF in the host or in the environment.17,19,20

Virulence factors, mechanisms and pathogenesis

Typical EPEC strains adhere to HeLa, HEp-2, and other cell lines and to organ cultures in vitro in a distinctive pattern of three-dimensional microcolonies, a so-called localized adherence (LA) pattern.6,21 A similar adherence pattern has been seen in tissue biopsies of EPEC-infected humans.22

The LA phenotype is mediated by the BFP,23 which also contributes to antigenicity, autoaggregation, and biofilm formation.23–27 An operon of 14 genes contained on the pEAF is necessary for BFP expression, with bfpA encoding the major structural subunit (bundlin)28 and being highly conserved among EPEC1 and EPEC2 strains.

The self-transmissible pEAF pMAR2 is found among strains of the EPEC1 lineage and contains an intact transfer region, unlike pB171, which is more common among EPEC2 strains.29,30 Besides the bfp gene cluster, encoding BFP,23 the pEAF carries the per locus, encoding the transcriptional activator called plasmid-encoded regulator (Per).29 Between pMAR2 and pB171, the bfp and per loci share 99% sequence similarity,30 and both BFP and PerA have been shown to contribute to virulence in human volunteers.24 Recent comparative genomics of the EAF plasmids from diverse EPEC phylogenomic lineages demonstrated significant plasmid diversity even among isolates within the same phylogenomic lineage.31

Typical EPEC have the ability to form tight, spherical, bacterial autoaggregates when grown in liquid culture.32 Like LA, autoaggregation requires BFP. Typical EPEC also form biofilms on abiotic surfaces under static conditions, or in a flow through continuous culture system, and a model of EPEC biofilm formation has been proposed.26 Mutagenesis analysis has identified adhesive structures such as the common type 1 pilus (T1P), antigen 43, BFP and the EspA filament (see below) as participants in bacterial aggregation during biofilm formation on abiotic surfaces.26

A hallmark phenotype of both tEPEC and aEPEC is the ability to produce AE lesions.33 This phenotype is characterized by effacement of intestinal epithelial-cell microvilli and intimate adherence between the bacterium and the epithelial cell membrane. Directly beneath the adherent bacterium, marked cytoskeletal changes are seen in the epithelial cell membrane, particularly the formation of an actin-rich cup-like pedestal at the site of bacterial contact. AE lesions are observed in model EPEC infections with cultured cells and mucosal explants, as well as in intestinal biopsies from EPEC-infected infants or animals.6

AE lesions are encoded by LEE, which is a ∼35-kb pathogenicity island (PAI)34 that is organized into five operons (LEE1 to LEE5).35–37 The LEE1, LEE2, and LEE3 operons encode components of a T3SS, and the global regulator Ler (LEE-encoded regulator).38 LEE4 encodes the T3SS-secreted proteins EspA, EspB, and EspD (EPEC-secreted protein), which are components of the translocation apparatus by which other effector proteins are translocated into the cell. LEE5 encodes the adhesin intimin and its translocated receptor, Tir.39

Intimin is a 94-kDa protein encoded by the eae gene and required for intimate adherence of EPEC to host cells at the sites of AE lesions.6 The N-terminus of intimin is highly conserved, whereas the C-terminus is highly variable.40 Differences in the C-terminus of intimin have been used as a basis for classification into several distinct subtypes, represented by the Greek letters α (alpha) through ζ (zeta)41,42; the α subtype is expressed by EPEC1 strains while subtype β is associated with human EPEC2 strains. The N-terminus of intimin anchors the protein in the EPEC outer membrane, whereas the C-terminus extends from the EPEC surface and binds to the Tir. Intimin–Tir interaction leads to intimate adherence and pedestal formation beneath adherent bacteria,39 and inhibits NF-κB activity through tumor necrosis factor alpha (TNF-α) receptor-associated factors.43 In addition to Tir, the EPEC genome contains six other LEE-encoded effector proteins that are translocated into the cell (Map, EspF, EspG, EspZ, EspH, and EspB), which interfere with different aspects of the cell's physiology.13,36,37,44

In addition to the LEE effectors, various non-LEE (Nle)-encoded effector genes (cif, espI/nleA, nleB, nleC, nleD, nleE, nleH)36,44 have been described, which are located outside the EPEC LEE region, in at least six chromosomal PAIs, or in prophage elements.45,46 The Nle proteins have been shown to disrupt the cytoskeleton and tight junctions of the host cell, and to modulate or prevent the host inflammatory response.45–47 Although they are not required for AE lesion formation, it is understood that they contribute to increased bacterial virulence.44

Intracellular tEPEC have been observed both in tissue culture and in small intestinal biopsies from an EPEC-infected infant.6 Two studies have reported that O111:NM strains contain plasmid sequences that confer invasiveness upon E. coli K12 strains containing the cloned fragments.48,49 Sequences homologous to these cloned genes are present in only a minority of tEPEC strains (Scaletsky et al., unpublished data).

Typical EPEC strains encode a large surface protein, lymphocyte inhibitory factor (LifA), which inhibits the expression of multiple lymphokines and inhibits lymphocyte proliferation.50 Two related genes efa1 and toxB have been implicated in adhesion to epithelial cells.51,52 There is evidence indicating that Efa1/LifA contributes to epithelial cell adherence in vitro53 and is required for intestinal colonization of mice by the related AE pathogen Citrobacter rodentium.54

Some tEPEC strains possess other fimbriae or pili in addition to BFP. Type 1 fimbriae of EPEC have been found to be antigenic in volunteer studies; however, they do not have a role in adherence to epithelial cells in vitro.6 In addition, some EPEC strains have conserved fimbrial genes encoding homologs of long polar fimbriae (LPF),55 but a number of polymorphisms within the lpfA genes have been identified.56 Initial studies have indicated that LPF is apparently not necessary for adherence and AE lesion in human biopsies.55 The E. coli common pilus (ECP) has also been shown to act as an accessory adherence factor in EPEC, playing a role during cell adherence and/or in bacterium-bacterium interactions.57 However, the significance of ECP to EPEC pathogenesis has not been determined. Interestingly, it has been shown that some tEPEC strains may produce a hybrid adherence phenotype in HeLa cells, i.e., LA and aggregative (AA)-like pattern concurrently (LA+/AA-like+).58 Recently, it was shown that at least some of these LA/AA-like+ strains bear large plasmids, distinct from the pEAF, that encode a so far unknown adhesin.59 It has been proposed that the ability of such strains in producing AE lesions and an AA-associated biofilm concomitantly could worsen the patient's clinical condition, leading to persistent diarrhea.59

Flagella may also be involved in tEPEC adherence to epithelial cells,60 since certain EPEC mutants are markedly impaired in their ability to adhere and to form microcolonies. Furthermore, in one study, purified EPEC flagella and anti-flagellum antibodies were both effective in blocking the adherence of several EPEC serotypes.60 However, another study could not confirm the role of flagella in EPEC adherence.61

Some tEPEC strains harbor the astA gene, which encodes the enteroaggregative E. coli heat-stable enterotoxin 1 (EAST1).62,63 A recent study reported that 11 of 70 (16%) tEPEC strains tested harbored an intact astA gene.64 Typical EPEC strains of serotype O86:H34 produce cytolethal-distending toxin (CDT).65 The significance of EAST1 and CDT toxins in EPEC pathogenesis remains unknown.

Autotransporter (AT) proteins, which have been associated with bacterial adherence, aggregation, biofilm formation, invasion, and toxicity66 in Gram-negative bacteria, have also been described among EPEC strains.67 One such protein, EspC, which is secreted by the type V secretion system and is injected by the T3SS in epithelial cells, has an IgA protease-like activity and, once in the host cytoplasm, has various cytopathic effects, including cytoskeletal damage,68 enhanced lysozyme resistance,70 hemoglobin degradation,69 hydrolysis of pepsin, factor V, and spectrin,70 and fodrin and focal adhesion protein degradation.71 In addition, oligomerization of EspC gives rise to rope-like structures that serve as a substratum for adherence and biofilm formation as well as protecting bacteria from antimicrobial compounds.72

A three-stage model of tEPEC adhesion and pathogenesis, consisting of LA, signal transduction, and intimate attachment with pedestal formation, was proposed.73 Simultaneously with intimate attachment, a series of bacterial effector proteins are injected into host cells, where they subvert actin polymerization and other host cell processes.37,44 In the earliest stage and under correct environmental conditions, tEPEC express BFP, intimin, and the T3SS/translocon apparatus. Next, EPEC adhere to the surface of the intestinal epithelium via BFP and EspA filaments, and the T3SS injects the bacterial translocated intimin receptor (Tir) and effector proteins (EspB, EspD, EspF, EspG, and Map) directly into the host cell.37 The effectors activate cell-signaling pathways, causing alterations in the host cell cytoskeleton and resulting in the depolymerization of actin and the loss of microvilli. Finally, bacteria intimately adhere to host cell by intimin–Tir interactions, causing a cytoskeletal rearrangement that results in pedestal-like structures. Tir promotes cytoskeletal reorganization through interaction with neural WASP (Wiskott-Aldrich syndrome protein) (N-WASP) and subsequent activation of the Arp2/3 complex,45 leading to the effacement of the microvilli and the production of pedestals.44,74 The translocated effectors disrupt host cell processes, resulting in loss of tight-junction integrity and mitochondrial function, leading to both electrolyte loss and eventual cell death.45

For actin dynamics subversion, tEPEC usually recruits Nck to the adhesion site in a Tir phosphorylated Y474-dependent mechanism. In turn, TirEHEC (enterohemorrhagic E. coli [EHEC] O157:H7) is devoid of an Y474 equivalent and employs EspFU/TccP (Tir-cytoskeleton coupling protein), a T3SS-translocated effector protein that binds N-WASP, leading to Nck-independent actin polymerization.45

aEPEC are devoid of pEAF and do not produce BFP. It is important to point out that EPEC strains of serotypes O128:H2 and O119:H2 contain a pEAF with defective bfp operons, which contain part of the bfpA gene but have the rest of the bfp gene cluster deleted. Thus, they are classified as aEPEC.12,75 Most aEPEC produce adherence patterns categorized as LA-like, with loosened microcolonies compared to those of the tEPEC LA pattern.12,76,77 In addition, some isolates express the aggregative (AA) or diffuse (DA) patterns of adherence, which are characteristics of the EAEC and DAEC pathotypes, respectively,20,78 or adhere in undefined patterns or are non-adherent.20,78–80 Remarkably, the epithelial cell adherence phenotype displayed by aEPEC is determined in prolonged assays (6h) of bacteria-cell interaction.12,76,77 In addition, it has been suggested that lack of the pEAF-encoded Per proteins in the regulatory cascade of the aEPEC virulence genes may promote delayed AE lesion formation, probably making it difficult for such strains to cause disease.81

The prevalence of intimin subtypes among aEPEC strains has been reviewed.18,67 Intimins classified as beta1, epsilon1 and theta appear as the most frequent among aEPEC.78,82–85 In addition, some aEPEC strains bear adhesive-encoding genes that have been originally described in other DEC pathotypes and/or in extraintestinal pathogenic E. coli.79,80,82,86–88 This observation suggests that aEPEC could employ additional adherence mechanisms besides the Tir-intimin interaction. The only adhesin first characterized in an aEPEC strain (serotype O26:H11) is the locus of diffuse adherence (LDA), which is an afimbrial adhesin that confers the diffuse pattern of adherence on HEp-2 cells, when cloned in E. coli K-12 strains.89 The T3SS-translocon has been also shown to contribute to the adherence efficacy of an aEPEC strain in vitro.90 The prevalence of these different adhesins among aEPEC has been recently reviewed.18,67,91

Moreover, it has been recently shown that the flagellar cap protein FliD of an aEPEC strain (serotype O51:H40) binds to unknown receptors on intestinal Caco-2 cell microvilli.92 Interestingly, an anti-FliD serum and purified FliD reduced adherence of the aEPEC as well as that of tEPEC, EHEC and ETEC prototype strains to the same cell line.92 Furthermore, it has been suggested that adherence of aEPEC of serotype O26:H11 may be mediated by binding of the flagellin protein FliC (the subunit of the flagella shaft) to cellular fibronectin.93 However, the role of the flagella in aEPEC in vivo colonization has yet to be investigated.

Atypical EPEC strains have also been shown to adhere to abiotic surfaces (polystyrene and glass).94,95 The non-fimbrial adhesin curli and the T1P have been shown to mediate binding to these surfaces in some aEPEC at different temperatures.90,96

The LEE region of some aEPEC strains display a genetic organization similar to that found in the tEPEC prototype E2348/69 strain.97 Although the T3SS-encoding genes are considerably conserved,97,98 the effector protein-encoding genes display important differences, and remarkable differences can be detected at the 5′ and 3′ flanking regions of aEPEC, suggesting the occurrence of different evolutionary events.99 Atypical EPEC strains may carry two tccP variants, tccP and/or tccP2, suggesting that some aEPEC strains may use both Tir-Nck and Tir-TccP pathways to promote actin polymerization.100 Interestingly, Rocha and colleagues101 showed that transformation of a non-adherent aEPEC strain (serotype O88:HNM) with a TccP expressing-plasmid, conferred this strain the ability to adhere to and to induce actin-accumulation in HeLa cells.

The occurrence and prevalence of Nle in aEPEC strains have been recently reviewed.67 It has been suggested that different isolates can employ distinct strategies to promote damage to the host and cause disease.45 In addition, the Nle effectors Ibe (invasion of endothelial cells) and EspT have been originally described and characterized in aEPEC strains.102,103 Ibe appears to regulate Tir phosphorylation and to enhance actin polymerization and pedestal formation,103 while EspT104 modulates actin dynamics, leading to membrane ruffling and cell invasion, and induces macrophages to produce interleukins IL-8 and IL-1β and PGE2.102

Invasion of epithelial cells in vitro in an intimin-dependent pathway has been described in an aEPEC strain,105 but further studies pointed out that the invasive phenotype is not a common characteristic among aEPEC.106 Despite their invasive potential in vitro,107 most aEPEC are considered extracellular pathogens.5

It has been shown that apical infection of cultured human mucin-secreting intestinal HT29-MTX cells by some aEPEC strains may induce increased production of secreted MUC2 and MUC5AC mucins and membrane-bound MUC3 and MUC4 mucins.108 This observation suggests that the apically adhering bacteria could exploit large amounts of mucins to grow more efficiently in the host intestines, characterizing a putative new virulence mechanism in aEPEC.108

AT proteins have also been shown to be produced by some aEPEC strains.67 Abreu and collegues109 have shown that the AT protein encoded by the ehaC gene, which is involved in biofilm formation in EHEC strains, was the most frequent, with a significantly higher prevalence than in tEPEC. Although the prevalence of the AT protein Pic (protein involved in intestinal colonization), formerly identified in EAEC, is not a common finding in aEPEC strains, it also appears to mediate colonization of mouse intestines, hemagglutination, mucin cleavage, and complement components degradation.110 More recently, some aEPEC strains were shown to cause cell damage by secreting the AT protein Pet (plasmid encoded toxin) to the extracellular environment.111

Epidemiology

The prevalence of EPEC infections varies between epidemiological studies on the basis of differences in study populations, age distributions, and methods (serotyping, adherence patterns, and presence of the eae or conserved LEE genes) used for detection and diagnosis.112 In addition, differences in geographic regions, periods of time and socioeconomic class may also contribute to differences in the epidemiology of EPEC-induced diarrheal disease.113 Lack of discrimination between tEPEC and aEPEC in some studies also makes such analysis difficult.

Diarrhea due to tEPEC decreases with age, and infections in adults are rarely reported. This apparent resistance in adults and older children has been attributed to the loss of specific receptors with age or development of immunity.6

For many decades, studies conducted worldwide have shown that tEPEC serotypes are strongly associated with diarrhea in children <1 year of age, mainly in poor children in urban centers.6,12,15 The association with diarrhea was particularly strong in infants less than 6 months of age. Studies in Brazil, Chile, Mexico, and South Africa, showed that 30–40% of infantile diarrhea cases were due to tEPEC serotypes.15,112,114 However, the epidemiology of EPEC infections has shifted. In numerous developing countries, where the prevalence of EPEC infection had been high until the 1990s, recent studies have not identified a significant association between tEPEC and infantile diarrhea. In Brazil, 92% of EPEC isolates collected from children between 2001 and 2002 were atypical,115 compared to 38% in a 1998–1999 study.79 However, other studies still report tEPEC being more prevalent than aEPEC as a cause of diarrhea.116 In addition, in some less developed areas (Africa and Asia), tEPEC are still some of the most important enteropathogens.117–122 Based on the recently completed Global Enteric Multicenter Study (GEMS) involving children less than 5 years of age from seven sites in Africa and Asia, tEPEC was significantly associated with moderate to severe diarrhea in children under 2 years of age in Kenya, whereas aEPEC was not associated with this type of diarrhea.118

Transmission of tEPEC follows a fecal-oral process through contaminated surfaces, weaning fluids, and human carriers.123 Although rare, outbreaks among adults seem to occur through the ingestion of contaminated food and water; however, no specific environmental reservoir has been identified.6 The infective dose in adult volunteers is high, at 108 to 1010 organisms,124 while the infective dose that causes disease in children is unknown. EPEC outbreaks have been reported to show a seasonal distribution with peaks during the warm months.6,125 Humans are the only known reservoir for tEPEC, with symptomatic and asymptomatic children and asymptomatic adults being the most likely source.6

In contrast to tEPEC, aEPEC have been found in diarrheic patients of all ages and in adults with HIV-AIDS.82,126–128 Furthermore, the proportion of aEPEC strains has increased, and aEPEC strains have outnumbered tEPEC strains and have also been associated with childhood diarrhea in some developing and developed countries.12,18,67,91,112,129 However, the increase in prevalence of aEPEC may also reflect the refined discrimination between tEPEC and aEPEC.12,18,91

The role of aEPEC in diarrhea is not clear because of its detection at similar rates in both diarrheic and non-diarrheic patients in various geographical areas.18,91,128 In studies conducted in the last five years, aEPEC have been found at rates varying from ∼0.05 to ∼12% in diarrheic versus 0 to ∼14% in non-diarrheic patients.67 Some recent studies have also implicated aEPEC as the cause of persistent and bloody diarrhea.18,91 Moreover, aEPEC strains have been associated with diarrheal outbreaks in Finland, United States, Japan, China18,91,112 and Brazil.85

In contrast to tEPEC, which are seldom found in animals,12 many aEPEC strains have been found in both diarrheic and healthy animals.18,67 Interestingly, animal aEPEC serogroups associated with human diarrhea have been identified (e.g., O26, O103, O119, O128, O142 and O157).18,130,131 Serotyping and molecular methods such as multilocus sequence typing (MSLT) and pulsed field gel electrophoresis (PFGE) have contributed to demonstrating that domestic and wild animals and the environment are potential sources of aEPEC for human infections in several regions.18,67,91,131 Therefore, although no direct transmission from animals to humans has been shown so far, it is reasonable to suggest that some aEPEC strains are potentially zoonotic pathogens, with a large variety of animal species serving as important reservoirs.67,91 In addition, foods including raw meat, pasteurized milk and vegetables and water have also been implicated as vehicles of aEPEC in human infections.67

aEPEC strains comprise a very assorted group with various additional virulence mechanisms that altogether can modulate the disease outcome or their occurrence in asymptomatic persons. There have been continuous advances in our knowledge of the genetic background and pathogenicity of aEPEC as well as in the information gathered from epidemiological studies, and may contribute to the discrimination between strains that cause diarrhea and those that cause asymptomatic infections.

Detection and diagnosis

EPEC can be detected by DNA probes or PCR assays using primers targeting the eae and stx genes.132,133 All eae-positive and stx-negative E. coli strains are further tested by PCR for the presence of the bfpA gene encoding bundlin6 and/or the EAF plasmid to differentiate tEPEC from aEPEC.134,135 However, this may fail to identify all bfpA-positive EPEC strains, since multiple alleles of bfpA have been identified,136 suggesting that some current PCR methods may fail to identify all bfpA-positive EPEC strains.

However, in routine microbiology laboratories, all E. coli colonies obtained from primary isolation plates are traditionally screened by slide agglutination assays using sera against the classical EPEC serogroups O26, O55, O86, O111, O114, O119, O125, O126, O127, O128, O142, and O158.137 This method is practical and easy to perform, the main advantage of which is the commercial availability of the sera. However, the disadvantage of this method is the heterogeneity of EPEC serogroups that can comprise categories other than EPEC, the inability to distinguish tEPEC from aEPEC within these serogroups, and the occurrence of EPEC strains belonging to serogroups other than the classical EPEC serogroups.12,18,138,139

Since EPEC strains are defined based on their virulence properties, a set of proteins, including intimin, BFP and T3SS secreted proteins can be considered targets for diagnosis. BFP expression has been considered the phenotypic marker of tEPEC.18,78,140 Immunofluorescence and immunoblotting tests using monoclonal or polyclonal antibodies against BFP have been employed.141,142 These cited authors detected the production of BFP on different media, in which they reported that 91% of the tEPEC strains tested produced BFP in Dulbecco's Modified Eagle Medium (DMEM), 89% in MacConkey, and 83% in EMB agars. These results are particularly interesting, since MacConkey and EMB agars are routinely used for the identification of lactose-fermenting E. coli isolated from diarrheal stools. A colony immunoblot assay for tEPEC detection based on BFP expression was also standardized using a rabbit tEPEC anti-BFP polyclonal serum. Standardization was done after growing the bacterial isolates on DMEM agar containing fetal bovine serum or tryptic soy agar containing 5% washed sheep blood (TSAB). This test showed a positivity of 92 and 83% and specificity of 96 and 97%, respectively, when the culture was done in DMEM and TSAB. This method combines the simplicity of an immunoserological assay with the high efficiency of testing a large number of EPEC colonies.140

Concerning intimin detection, a rabbit polyclonal sera raised against the conserved region of intimin (Int388-667)143 was employed in order to detect tEPEC isolates expressing α, β, γ, δ and ¿ intimin reported an application of immunoblotting with 100% specificity and 97% sensitivity in the detection of eae positive E. coli strains.144–146 These authors clearly demonstrated that polyclonal rabbit antisera is suitable for immunoblotting as a diagnostic tool, and showed that protein denaturation and linearization is a critical step for anti-intimin antibody accessibility. Indeed, even employing the recombinant antibody such as single chain fragment variable (scFv-intimin),147,148 merely by immunofluorescence the scFv-intimin was able to detect tEPEC, aEPEC, and EHEC isolates, showing that intimin can be a target for EPEC and EHEC diagnosis after bacterial permeabilization.148

Regarding secreted proteins, Lu et al.149 developed a new practical method to identify EPEC by detecting the E. coli secreted protein B (EspB) in the culture supernatant by reversed passive latex agglutination (RPLA), after the strains have been cultivated in DMEM. In addition, Nakasone et al.,150 established a rapid immunochromatographic (IC) test to identify the presence of EspB in EPEC and EHEC isolates. The detection limit of the test has been reported to be 4ng/mL, and the results showed 96.9% sensitivity and 100% specificity. The IC test for the detection of EspB may be a practical method to define EPEC or EHEC both in clinical laboratories and the field.150

In addition, a rapid agglutination test using latex beads coated with anti-EspB mAb was standardized, showing 97% sensitivity, 98% specificity and 97% efficiency, which is required for the diagnosis of enteropathogenic diseases and can be employed in developing countries with poorly equipped laboratories.151

Enterohemorrhagic (Shiga toxin-producing) E. coli (EHEC/STEC)

EHEC/STEC represent a well-known group of foodborne pathogens distributed worldwide. The ability to produce one or more of the Shiga toxin (Stx) family cytotoxins152 constitutes the main virulence attribute of this pathogroup of E. coli. A wide array of infections from mild and almost unapparent diarrhea to more serious manifestations such as hemorrhagic colitis (HC) and the development of a life-threatening syndrome known as hemolytic uremic syndrome (HUS) are caused by EHEC/STEC. Infants and children are the main affected patients, and although the incidence of infection varies in different regions, the impact and importance of EHEC/STEC infections in public health is immense, being the main cause of acute renal failure in children in many countries. The perspective of EHEC/STEC infections has been previously described,153,154 but a considerable amount of information has been obtained in more recent years related to the epidemiology, ecology and virulence properties of these bacteria.

E. coli O157:H7 serotype was the first to be linked to HC and HUS cases in the early 1980s, and has been since then responsible for numerous outbreaks and sporadic cases of severe diseases all over the world, therefore considered to be the prototype of this pathogenic group of bacteria.155 It is well known that hundreds of other E. coli serotypes can harbor the stx genes, but epidemiological studies carried out worldwide have proven that only some of them have been responsible for causing human diseases. Some serogroups including O26, O45, O103, O111, O121 and O145 can be highlighted among those most commonly related to human infections.156 Moreover, in recent years the emergence of some particular clones such as the hybrid O104:H4 enteroaggregative E. coli carrying Stx2 genes, responsible for a severe outbreak of HUS starting in Germany in 2011,157 the spread of a new O26:H11 clone in Europe,158 and some other hybrid clones,159 suggests that the mobility of genes and certainly the host background are important features implicated in their pathogenic potential.

Virulence factors, mechanisms and pathogenesis

The common feature among EHEC/STEC isolates is the ability to produce Stx. This family of toxins has a conserved AB5 subunit structure, composed of one active A subunit linked to a pentameric B subunit responsible for the binding of the toxin to specific glycolipid receptors on the surface of target cells. The stx operon is usually found within the sequence for an inducible, lysogenic, lambda-like bacteriophage. Stxs inhibit protein synthesis by removing an adenine residue from the 28S rRNA of the 60S ribosome.152 However, besides this activity, studies have described that Stx also acts on cell signal transduction and immune modulation causing proinflammatory and pro-apoptotic responses.159 Two major families, Stx1 and Stx2, have been recognized, and on the basis of sequence diversity, each is composed of several variants. The Stx1 family is more homogenous and includes Stx1a, Stx1c and Stx1d; while the heterogenous Stx2 group is composed of Stx2a, Stx2b, Stx2c, Stx2d, Stx2e, Stx2f, and Stx2g.160 It should be mentioned that the association of some variants such as Stx2a, Stx2c or Stx2d with HC and HUS has been highlighted compared to some others that seemed to be more related to uncomplicated cases of diarrhea such as Stx1variants or even Stx2e, Stx2f and Stx2g, which are uncommonly found causing human infections so far.161,162 Indeed, the higher association of Stx2 with severe diseases has been extensively studied by using Vero and endothelial cell lines as well as some animal models.159 Moreover, knowledge of stx phage characteristics and behavior has helped our understanding of how differences in expression of Stx between EHEC/STEC isolates may contribute to pathogenesis and disease.163

The ability to adhere to intestinal epithelial cells is another key event in EHEC/STEC pathogenesis. The presence of the chromosomal pathogenicity island LEE,164 also present in isolates belonging to the EPEC pathotype, is common. Although LEE has been described in the major EHEC/STEC serotypes responsible for a high proportion of HC and HUS cases in several countries, its presence is not a required condition for the occurrence of more serious infections as initially thought, because some LEE-negative strains are also capable of causing outbreaks and sporadic cases of HUS.165,166

Therefore, it is clear that EHEC/STEC pathogenesis is a multistep process, and besides the production of Stx toxins and the AE lesion, other factors including different types of toxins and adhesins have been described and found to be involved in virulence.159

One should also consider that as a pathogen of the human gastrointestinal tract the ability of EHEC/STEC to monitor nutrients in the gut milieu, and translate this information to sense the host physiological state in order to program the expression of its virulence markers has a pivotal role on the development of infection.167 In addition, it has been shown that EHEC/STEC can also cross-communicate with the host by exploiting the autoinducer-3 (AI-3)/epinephrine/norepinephrine signaling system to express two important virulence traits, motility and A/E lesion, required at different time points during intestinal colonization.168

The ability to adhere, colonize and form biofilm on food and several types of surfaces may be a way to be an important source and/or vehicle of transmission of EHEC/STEC. In addition, biofilm may also act as bacterial protection against adverse environmental conditions. A study conducted by Biscola et al.,169 evaluated the capacity of biofilm formation in EHEC/STEC strains isolated from different reservoirs and serotypes. The authors observed that the ability to adhere to abiotic surfaces forming biofilms, under defined culture conditions, occurred in an array of wild-type O157 and non-O157 strains. Biofilm production was identified in several non-O157 STEC serotypes of human, animal, and food origin. On the other hand, among the O157 strains, only those isolated from the animal reservoir and from a water sample produced biofilm. A close correlation between biofilm formation and expression of curli fimbriae and cellulose was observed among O157 strains. However, in addition to curli, the presence of other factors such as type 1 fimbriae and AT proteins may be associated with the ability to form biofilm in non-O157 strains. Matheus-Guimarães et al.,170 studied O157 and non-O157 EHEC/STEC strains isolated from bovine hides and carcasses and showed that different sets of genes were involved in the interactions of the bacteria with biotic and abiotic surfaces. Moreover, the detection of an O157 strain that was able to form biofilm on both glass and polystyrene and that adhered to and invaded human cells, suggests an important ability of this isolate to persist in the environment and interact with the host. In fact, cell invasion and survival of some EHEC/STEC strains in cultured human intestinal epithelial cells has been previously described.171 It should be mentioned that this invasive characteristic has been identified in some EHEC/STEC serotypes, many of which are responsible for human infections.170–173 Therefore, it is conceivable that this virulence strategy may help bacteria to overcome host defense mechanisms and certainly contributes to their persistence in the zoonotic reservoir, ensuring efficient environmental and food transmission.

Another topic of interest has been the analysis and comparison of the virulence profile of EHEC/STEC strains isolated from the animal reservoir and environment with strains recovered from human infections. In general, these studies have shown that despite serotype diversity, the stx subtypes and the virulence profile identified among isolates from the animal reservoir and environment are similar to the isolates recovered from patients.173,178–180 There has been particular interest in some STEC serotypes that have been responsible for causing severe human infections, such as O113:H21, but unlike others, they do not produce adhesins encoded by LEE. By using a PCR microarray, 41 virulence or genetic markers were tested in a panel of 65 O113:H21 strains isolated from clinical infections, environment and food from various countries.174 The results obtained showed no clear differences in these genetic markers between the pathogens recovered from HUS cases and the environmental strains. Moreover, only stx subtypes associated with human infections were identified in all isolates, therefore suggesting that the environmental isolates have the potential to cause human diseases.

Epidemiology

The incidence of HUS cases in Brazil is low,175 and although some hypothesis has been proposed to explain this fact, there are limited data on the immune response against Stx. In an attempt to overcome this gap, prevalence of anti-Stx2 antibodies in sera of children diagnosed with HUS and of healthy children was recently determined.176 The percentage of individuals showing antibodies against Stx2 was higher among HUS patients than controls, and the results also confirmed that STEC strains are circulating in our settings despite the low number of identified HUS cases.

Among the several serotypes associated with human infections, O157:H7 is responsible for more severe cases. Epidemiological investigations of diarrheal outbreaks conducted in four Brazilian states showed that O157:H7 strains were isolated from two hospitalized patients, one with HUS and the other with bloody diarrhea.177 Besides, O157:H7, EHEC/STEC strains belonging to the top six most important non-O157 serogroups such as O26, O103, O111 and O145 were identified, all of which were recovered from ambulatory patients. In addition, some uncommon serogroups including O1, O24 and O77 among others were also detected, but they were all associated with acute diarrhea. It is interesting to note that the majority of patients from whom STEC was isolated were female (57%), and that patients’ ages ranged from 8 months to 80 years, with most being less than five years old (54%).177

The distribution of EHEC/STEC in the gastrointestinal tract of a wide variety of animals indicates the zoonotic character of its infections. The role of different animal species as asymptomatic carriers of EHEC/STEC has been extensively studied in the last years in Brazil. Besides cattle, which are their most common natural reservoir,173,178 the presence of these pathogens has been identified in the feces of dairy buffaloes,179 sheep,180,181 pigs,182,183 birds,184 and fishes.185 It is noteworthy that some relevant serotypes linked to human infections such as O103:H2 and O157:H7 have been recovered from the feces of sheep186 and cattle173 respectively. Additionally, the high prevalence of O157:H7 EHEC/STEC strains identified in hides of cattle sent to slaughter in a Brazilian processing plant178 certainly represents a relevant issue that should be considered when thinking about interventions targeting EHEC/STEC related to animal handling, from farm to slaughter, as well as the implementation of food safety throughout production and processing.

The presence of EHEC/STEC in the environment is another issue of concern, since they can survive in the soil, manure, pastures and water, which thus represent important vehicles of transmission. The isolation of STEC strains from drinking water supplies, collected in different municipalities in northern Paraná State, has been recently described, highlighting the importance of drinking water, especially that from untreated water supplies, as a source of STEC strains potentially pathogenic for humans.187 Taking into account that chicken litter is very useful as an organic soil fertilizer for the production of fruits and vegetables in our settings, the detection of STEC in organic chicken fertilizer used on farms188 also represents a significant public health safety hazard.

Although data on the detection of EHEC/STEC in foods in Brazil are still scarce, the isolation and identification of O157:H7 serotype from a ground beef sample was described for the first time,189 while O125:H19 and O149:H8 STEC serotypes were found in refrigerated raw kibbe collected from retail establishments.190 On the other hand, EHEC/STEC has not been detected in pasteurized cow's milk samples collected in dairies in northwestern Paraná State191 or in raw milk, pasteurized milk, Minas Frescal cheese and ground beef samples collected in Minas Gerais.192 One should be aware that despite difficulties in the detection and isolation of EHEC/STEC from foods, the implementation of the most sensitive methods in most laboratories should be the main goal in the near future to help in the analysis of the risk posed by foods as vehicles of STEC transmission to humans.

Detection and diagnosis

An important concern is how to detect Shiga toxin-producing strains either in stools of infected patients or contaminated food, since selective enrichment is necessary.193,194 For routine diagnosis, some protocols have already been described.139 However, the gold standard for Stx detection is still the evaluation of the cytotoxicity of bacterial culture supernatants to eukaryotic cells.195,196 Thus, multiplex PCR including stx gene and other virulence genes could be useful in screening for STEC using bacterial confluent growth zones or sorbitol fermenting and non-fermenting colonies taken from SMAC.197

Numerous assays for the diagnosis of STEC have been developed on the basis of the detection of Stx1 and/or Stx2, which represents the major virulence factors of this E. coli category.198 Sensitivities and specificities vary according to the test format and the manufacturer.199–205 Nevertheless, the standard by which each manufacturer evaluates its tests also varies; therefore, a direct comparison of performance characteristics of various immunoassays has not been performed.198,206,207 Moreover, these commercially available tests are not affordable for developing countries. Thus, to outline this, previous works have established different formats of immunoassays, employing either a mixture of rabbit anti-Stx1 and anti-Stx2 sera by indirect ELISA or polyclonal and monoclonal antibodies in a capture ELISA assay for the detection of STEC.207–209 The standardized methods are reproducible, fast, easy to perform, showing high sensitivity in detecting Stx by capture ELISA, even in low-producing isolates. These assays have not yet been evaluated in terms of industrial quality control and commercial availability, but the estimated cost of the assay is around US$70 per 96 detections, which is realistically inexpensive for developing countries.

These monoclonal antibodies were rebuilt resulting in single chain fragment variable (scFv) fragments. Stx2-scFv was obtained from a bacteria-induced culture and showed diagnostic ability; the scFv fragment was able to recognize the majority of Stx2-producing strains, with 79.3% sensitivity (confidence interval of 60.3 to 92%), and no reactivity was observed with the non-producing strains, indicating as high as 100% specificity (confidence interval of 86.8–100%).210 It is worth mentioning that none of the commercially available immunoenzymatic tests for Stx1/2 toxin detection employ recombinant antibodies produced in bacteria, which indeed will reduce the costs of the diagnostic assays.198

Enteroaggregative E. coli

EAEC is the diarrheagenic E. coli pathotype defined by showing the characteristic AA pattern on epithelial cells in culture.211 The AA pattern was defined in 1987 when Nataro et al.,212 distinguished the previously described “diffuse adherence” as the truly diffuse adherence (DA) and the AA pattern. The standard AA was characterized by adherent bacteria in a stacked-brick arrangement on the surface of epithelial cells and also on the coverslip between cells. Strains displaying the AA pattern were then categorized as “enteroadherent-aggregative E. coli” but afterwards the category was called enteroaggregative E. coli or EAEC, the current nomenclature. The detection of AA in vitro is still the gold standard test to define EAEC; however, as described before, the AA pattern may be found in strains of other DEC pathotypes, such as aEPEC. Therefore, an up-to-date definition of EAEC is the diarrheagenic E. coli that produce AA in cultured epithelial cells but lack the main genetic markers that define other DEC pathotypes (EPEC, ETEC, EHEC, EIEC). An exception for that is the hybrid EAEC/STEC strain responsible for a massive outbreak of diarrhea and HUS in 2011 in Europe.213 This strain consists of an EAEC strain that acquired the Stx2-encoding phage. Therefore, this specific O104:H4 strain is a Stx-producing EAEC.

Diarrhea caused by EAEC is watery, often with the presence of mucus, with or without blood and abdominal pain, vomiting and low fever. Acute self-limiting diarrhea is the usual pathology, but some patients may develop protracted diarrhea, i.e., lasting more than 14 days.214 Prolonged diarrhea occurs depending on the host's immunity, nutritional status and genetic susceptibility.215 Genetic susceptibilities associated with EAEC diarrhea were identified in North American travelers to Mexico. Single nucleotide polymorphisms (SNP) in the IL-8 gene promoter and the promotor regions of the genes encoding lactoferrin, CD14 and osteoprotegerin as well were recognized as indicators for symptomatic EAEC infection.216–219

A well-described characteristic of EAEC strains is their heterogeneous nature when serotypes, genetic markers of virulence and phylogenetic groups are analyzed.220–225 This indicates that only EAEC strains carrying specific virulence factors are able to cause diarrhea. While these factors are unknown, some studies have demonstrated the association of specific virulence genes with diarrhea, such as pet or aafA in Brazil226 and sepA in Mali.223

Virulence factors, mechanisms and pathogenesis

Most of our knowledge about EAEC pathogenesis is based on data accumulated from studies with EAEC strain 042, since its association with human diarrhea in a volunteer study.227 These putative virulence factors include adhesins, toxins and secreted proteins. However, none of these factors are found in all EAEC strains.

The majority of these virulence factors are plasmid borne, including those mediating AA. Consequently, these high-molecular-weight plasmids are called pAA.220 Baudry et al.,228 developed a genetic probe (CVD432) for EAEC diagnosis on the basis of a fragment from pAA1 present in EAEC strain 17-2. In EAEC 042, many putative virulence factors are present in pAA2.220

Recently, a division of EAEC strains into typical or atypical subgroups was proposed. This classification is based on the presence or absence of aggR, a gene that encodes a global regulator of EAEC virulence genes.229 Therefore, it has been proposed that typical EAEC have more pathogenic potential by the presence of the AggR regulon and, consequently, pAA virulence factors.230 However, at least two outbreaks of diarrhea were caused by atypical EAEC,231,232 and atypical EAEC are commonly isolated from children with diarrhea, in some cases more frequently than typical strains.233,234

Numerous adhesins, cytotoxins, enterotoxins and secreted proteins have been characterized in EAEC strains since this pathotype definition.211,214

The most studied adhesins are the aggregative adherence fimbria (AAF/I-AAF/V) family, which includes five types.235–239 They mediate the AA pattern and biofilm formation. Afimbrial adhesins have also been characterized in EAEC strains, including outer membrane proteins between 30 and 58kDa.240–242 However, it has been shown that these structures are present in low frequencies in EAEC collections from different settings.221,226,243–245

Located in pAA2 of EAEC 042 is the aap gene, encoding an antiaggregation protein called dispersin.246 This protein is secreted and linked to lipopolysaccharide, neutralizing the negative charge of the bacterial surface leading to AAF projection and consequent dispersion along the intestinal mucosa.247 Although immunogenic, dispersin is found in other E. coli pathotypes and in commensal E. coli.248

Various toxins have been described in EAEC in association with the cytotoxic or enterotoxic effects of culture supernatants in vitro. The heat-stable toxin enteroaggregative E. coli heat-stable enterotoxin 1 (EAST-1) was the first toxin characterized in the EAEC pathotype.249 EAST-1 activates adenylate cyclase inducing increased cyclic GMP levels, effects observed in a Ussing chamber with rabbit ileum.250 ShET1 is an A:B type toxin that causes accumulation of fluid in rabbit ileal loops and has secretory response in Ussing chamber assays.251,252

The two AT proteins characterized in EAEC 042, Pet and Pic,253–254 are members of the serine protease autotransporters of Enterobacteriaceae, or SPATE.255 Pet is a cytotoxin that modifies the cytoskeleton of enterocytes, leading to rounding and cell detachment. The cytotoxic mechanism of Pet arises from the degradation of α-fodrin, a membrane protein of the enterocytes.256 Pic is a multitask protein that mediates hemagglutination, mucus cleavage and hypersecretion, intestinal colonization in mice, cleavage of surface glycoproteins involved in leukocyte trafficking and cleavage of key complement molecules.257,258 The phenotypes identified for Pic suggest its role in promoting colonization of the intestine and immune system evasion. SPATEs are immunogenic proteins, as evidenced by the presence of serum antibodies against Pet and Pic in children recovering from diarrhea caused by EAEC.259

In the years that followed the definition of EAEC as a pathotype, research in the field was dedicated to prove the pathogenic capacity of EAEC using different animal models260–262 and human volunteers receiving oral inoculum of different EAEC strains.227,235,263 Not all volunteers developed diarrhea after ingestion of different EAEC strains, the first evidence that strains of this pathotype are heterogeneous. Among the strains tested, EAEC 042 (serotype O44:H18) caused diarrhea in three out of five volunteers.227 Since then, strain 042 has been considered the prototype EAEC strain and is certainly the most studied strain of the pathotype.264 EAEC 042 was isolated from a case of acute infantile diarrhea in Peru.265 The clinical data obtained from the volunteers who developed diarrhea suggested that EAEC 042 caused secretory diarrhea, with abundant presence of mucus and absence of blood in the stool.

Studies employing different EAEC strains interacting with intestinal cells from animals or humans have been performed to elucidate the pathogenesis of this pathotype. Data from these in vitro, in vivo and ex vivo experiments strongly indicate that EAEC can bind to jejunal, ileal and colonic epithelium in the characteristic aggregative pattern, forming a strong biofilm in a mucus layer, followed by cytotoxic and proinflammatory effects.260,266–270 Fragments from terminal ileum and colon excised from pediatric and adult patients were incubated with EAEC strains that were capable to colonize the ileal and colonic mucosa in the typical stacked-brick pattern over an augmented mucus layer.270

All these lines of evidence in combination with the identification of several putative virulence factors in prototype EAEC strains allowed the proposal of a three-stage model of EAEC pathogenesis: (a) abundant adherence to the intestinal mucosa, (b) production of cytotoxins and enterotoxins, and (c) induction of mucosal inflammation.211 In the first stage, the contribution of fimbrial and afimbrial adhesins as well as other adhesive structures is essential. Several colonization factors have been identified in EAEC strains.271 In this stage, a characteristic increased secretion of mucus on the intestinal mucosa leads to the formation of a strong biofilm where EAEC are embedded.234,266,272 In the following step, EAEC produce cytotoxic effects on the intestinal mucosa due to the secretion of toxins, inducing microvillus vesiculation, enlarged crypt openings, and increased epithelial cell extrusion.266,273 EAEC-induced inflammation results from the strong colonization of the intestinal mucosa; however, all bacterial factors that contribute to this condition have not been identified. Inflammatory markers such as IL-8, IL-1β, interferon (INF)-γ and lactoferrin have been detected in stools of children and adults colonized by EAEC.274–276 Although this model summarizes the data so far obtained using in vivo, in vitro and ex vivo approaches it may not be valid for all strains.

A large foodborne outbreak of bloody diarrhea and HUS occurred in 2011 in Europe, affecting more than 4000 patients, most of them from Germany. This outbreak was caused by a Stx2-producing E. coli strain belonging to the serotype O104:H4. The genome of that strain was rapidly sequenced, revealing a unique hybrid combination of EAEC and STEC, i.e. the EAEC strain Ec55989 harboring the Shiga toxin 2-encoding prophage.166,213 Several virulence factors of typical EAEC are present in that strain, including AggR, dispersin, Pic and ShET-1. Also expressed are two Shigella autotransporter proteins called SigA and SepA, implicated in mucosal damage and colonization.277,278 Interestingly, the EAEC Ec55989 is the prototype strain for AAF/III.237 Conversely, the outbreak hybrid strain produces AAF/I, showing that the outbreak EAEC/STEC acquired an AAF/I-encoding plasmid.166,279 It has been proposed that the presence of these virulence factors combined is responsible for the highly virulent attributes of that strain.166,213

Epidemiology

EAEC is an emerging pathogen affecting children and adults worldwide, responsible for cases of acute and persistent diarrhea. Nevertheless, the most important impact in terms of morbidity is among children younger than 5 years living in developing countries.214 A meta-analysis study of the literature on the epidemiology of diarrhea that included the search of EAEC showed a statistical association of EAEC with acute and persistent diarrhea in developed and developing countries, with diarrhea in HIV-infected patients in developing countries, and adult traveler's diarrhea.280 In another meta-analysis study EAEC was associated with acute diarrhea in children living in South Asian countries.281

It is important to mention that data on the epidemiology of EAEC infection are somewhat inconsistent due to large variation in terms of method of detection, geographical location and patient age and socioeconomic status. Nonetheless, EAEC has been systematically identified as an emerging enteropathogen, strongly associated with acute and persistent diarrhea in children of developing countries. Moreover, in developed countries, EAEC have been frequently isolated from cases of diarrhea in children and adults in the last years.282,283

In addition, several foodborne outbreaks of diarrhea caused by EAEC have been reported in Europe, Japan, Mexico and India.231,232,284–286 One of them affected 2697 school children in Japan, after consumption of school lunches.232

Several studies have implicated EAEC as the predominant agent of persistent diarrhea in children.287–289 EAEC-mediated persistent diarrhea has been linked to malnutrition and decrease in physical and intellectual development in several studies from Brazil.274,288,290 Notably, asymptomatic patients infected with EAEC also exhibit growth retardation.274 Since its definition as a pathotype, high rates of asymptomatic young children carrying EAEC have been reported in several studies, involving subjects with low socioeconomic status in developing countries.214 The persistence of EAEC may induce chronic intestinal inflammation, even in the absence of diarrhea, reducing its absorptive function and leading to malnutrition.274,291 Growth impairment has also been observed in a mouse model of EAEC oral infection.292 Considering the high number of asymptomatic EAEC-colonized children in low-income countries, this pathotype has an important impact on public health as one cause of impaired physical and cognitive development.

EAEC is transmitted by the fecal-oral route by food or contaminated water.232,285,286 EAEC were detected in milk samples from infant feeding bottles that were handled by mothers with low socioeconomic status.293 Also, viable EAEC were isolated in tabletop sauces from Mexican restaurants.294 No relationship has been found between EAEC strains isolated from humans and different animal species, indicating that animals may not represent a reservoir of human pathogenic typical EAEC.295

EAEC has also emerged in the last years as an agent of urinary tract infections (UTI). Initially, Abe et al.,296 described the presence of EAEC virulence markers in strains isolated from UTI, which was subsequently observed by others.297–300 Also, the presence of uropathogenic E. coli (UPEC) markers in EAEC collections has been reported.301,302 These findings pointed out the potential for some EAEC strains to cause UTI.

A community acquired UTI outbreak caused by an EAEC strain of serotype O78:H10, occurred in Denmark.303 This multiresistant strain belonged to the multilocus sequence type ST10 and phylogenetic group A. This was the first time that EAEC was implicated as an agent of an outbreak of extraintestinal disease. The uropathogenic properties of this EAEC strain were conferred by specific virulence factors, such as the AAF/I fimbriae.304 Recently, EAEC was implicated as a causative agent of one case of urosepsis.300

Detection and diagnosis

Among the DEC pathotypes, EAEC is the most difficult to categorize, since it is a very heterogeneous group. The defining characteristic of EAEC is the AA pattern in human epithelial cells or on a glass substrate in a distinctive stacked-brick formation. Thus, the gold standard method for distinguishing EAEC is to culture five E. coli colonies per patient in static Luria-broth at 37°C, and then to infect semi-confluent HEp-2 cells for 3 or 6h, looking for the typical AA pattern.212,305 However, this test requires specialized facilities and is time-consuming, restricting its use only to research and certain reference laboratories.

Furthermore, despite that several protein components such as Pic, ShET1, EAST-1, and Pet are involved in the virulence of EAEC, none of them is present in all isolates. The presence of Pet in EAEC isolates was initially detected by immunoblotting assays after a preliminary step of culture supernatant concentration.256 Vilhena-Costa et al.306 developed a slot blot immunoassay that avoids the concentration step, allowing the detection of Pet directly from EAEC supernatant, after growing the EAEC bacterial isolate in TSB at 37°C for 4h. In this method, it was possible to evaluate Pet expression with specificity and reproducibility, using a rabbit polyclonal anti-Pet serum, which showed no cross-reaction with supernatants of non-Pet-expressing isolates and commensal E. coli.

Considering these difficulties, DNA probes were included as a valuable tool for EAEC detection.307 After sequencing the EcoRI-PstI fragment of pCVD432 (AA or EAEC probe) developed by Baudry et al.,228 primers complementary to this probe for PCR amplification were designed.308 This PCR assay was found to be a rapid, simple, and highly sensitive method, and therefore considered to be useful for screening stool specimens for the presence of EAEC strains. Rapid and practical multiplex PCR assays targeting more genes (aggR, aap and aatA, encoding the AggR regulator, dispersin and an ABC secretion system outer membrane protein, respectively) or aggR, pic and astA, encoding AggR, Pic and an EAST-1) have also been employed to detect EAEC strains.309–311 Monteiro et al.248 used PCR to evaluate aggR, aatA and aap in a collection of E. coli strains and found that aggR and aatA were more specific to EAEC than aap, suggesting that the simultaneous detection of aggR, aatA, and aaiA (a type VI secretion system protein) could be an improvement in the PCR detection of EAEC.

All these proposed PCR-based protocols detect plasmid genes, which disfavors the detection of atypical EAEC strains.305,309,312 Others, employing plasmid and chromosome loci, have not reported sensitivity and specificity of the assay.226,313,314 However, a multiplex PCR based on two genes encoded in the plasmid and two chromosome-borne genes is recommended to increase the ability to detect both typical and atypical EAEC strains. The aggR and aatA genes309,313 and aaiA and aaiG genes315 incorporated in the assay detecting aaiA, aaiG, aggR and aatA demonstrated 94.8% sensitivity and 94.3% specificity, and the assay was able to effectively detect both groups of EAEC among E. coli isolated from stool cultures.316 This method should improve EAEC detection, since this pathotype is responsible for acute and persistent diarrhea in children and adults and is also associated with foodborne diarrheal outbreaks.

Enterotoxigenic E. coli

ETEC strains are characterized by the production of colonization factors (CFs) and at least one of two enterotoxins: LT and ST. ETEC represents one of the most common causes of diarrhea in children in developing countries and in travelers to these regions. ETEC is also an economic burden to farmers and industry, where it is an important pathogen for broilers, swine, cattle and other farm animals. The group represents a highly diverse pathovar of diarrheiogenic E. coli, harboring mobile genetic elements such as plasmids and phages. ETEC heterogeneity was first demonstrated by phenotypic traits including the large diversity of lipopolysshacaride (LPS) and flagelin composition and the expression of different CFs and toxin types.317,318 Serological typing of ETEC strains have relied on the composition of outer membrane proteins and, mainly, in the somatic LPS (O) and flagellar (H) antigens.318–320 ETEC comprise more than 100 somatic serogroups (O) and at least 34 flagellar types (H), combined in an unpredicted number of O:H serotypes, but only a limited number of serotypes are associated with infectious diseases, such as O8:H9, O6:H16, O78:H12 and O25:H42, and are therefore of major clinical relevance.318,321

The genetic diversity of ETEC has also been evaluated by molecular approaches including random amplification of polymorphic DNA (RAPD), MLEE, PFGE, multilocus sequence type (MLST) and whole-genome sequencing.322–330 More recently, 362 human-derived strains were subjected to next-generation whole-genome sequencing; 21 genotypes could be identified, and ETEC strains could be classified into 5 major phylogroups (A, B1, B2, D and E).330 Genetic analyses demonstrated that clonally related ETEC lineages sharing the same serotypes and CF and toxin profiles have worldwide distribution.327,328,330–332 On the other hand, genetically distinct ETEC strains, frequently found among asymptomatic subjects show high antigen heterogeneity with regard to virulence traits and serotypes.333 Apparently, these strains have recently acquired the genes encoding virulence-associated traits, and their maintenance is driven by selective pressure.328,330

Virulence factors, mechanisms and pathogenesis

Following the initial discovery of the association of ETEC with diarrheic disease in humans in the 1950s, there was an intense effort to identify ETEC virulence-associated traits that could help to understand the physiology of the pathological process and lead to the development of specific diagnostic methods. ETEC strains characteristically produce adhesins, or CFs, proteinaceus complex that may take the shape of fimbrial, fibrillar or nonfimbrial structures on the bacterial surface. The adhesins expressed by ETEC strains facilitate the adherence of the bacteria to the intestinal mucosa and confer host specificity to the different strains.317,321

Approximately 30 antigenically distinct CFs have been identified in clinically relevant ETEC strains, but only a few are usually found among samples collected from diarrheic patients.317,330 Besides differences regarding biogenesis and structural organization, ETEC CFs show specific antigenic, genetic and biochemical features, which are currently used to cluster them into three main groups: the colonization factor antigen I (CFA/I)-like group, the coli surface antigen 5 (CS5)-like group and the class 1b group.317,334,335 The CFA/I-like group harbors the first described CF (CFA/I) and some of the most clinically prevalent CFs, including CS1, CS2, CS4, CS14, CS17, CS19 and putative colonization factor O71(PCFO71), while the CS5-like group comprises only CS5 and CS7. The class 1b group includes CS12, CS18, CS20 and the recently described CS26-28 and CS30 types.317,334,335 Additionally, genetic relationships are also observed between strains expressing CS8 and CS21, CS13 and CS23, as well as between strains expressing CS15 and CS22.336–338 Other previously characterized CFs, such as CS3, CS6, CS10 and CS11, are not classified into the known CS families.317 Some CFs, such as CS18 and CS20, are related to swine-derived ETEC fimbriae, which show a lower heterogeneity than those found in strains isolated from humans.317,321,339 Strains expressing CFA/I, CFA-II (CS1/CS3, CS2/CS3 or CS3), CFA-IV (CS4/CS6, CS5/CS6 or CS6), CS17 and/or CS21 are the most prevalent CFs found in epidemiological studies, whereas other CFs are found in ETEC strains not clearly linked to diarrheal disease.317,318,331

After adherence to the intestinal mucosa, ETEC strains produce enterotoxins, which are recognized as the second component associated with diarrheal disease. Two major categories of enterotoxins have been identified among ETEC strains, isolated either from humans or other animal hosts: LT and ST. Both toxin types mediate deregulation of membrane ion channels in the epithelial membrane, leading to the loss of ions and massive amounts of water, the major characteristic of watery diarrhea caused by these bacterial strains.340

LT are composed of five identical monomers (11.5kDa) arranged in a ring shape to form a pentameric B subunit, and a 28-kDa A subunit linked to the B subunit by the helical A2 domain. The B subunit binds to cell surface receptors, particularly to gangliosides, promoting toxin internalization and retrograte transport up to the endoplasmic reticulum, where the A1 domain is cleaved from the A2 domain and released to the cytoplasm. The A1 domain transfers the ADP-ribose moiety from the NAD+ cofactor to stimulatory G protein, which becomes active and capable of stimulating adenylate cyclase, leading to an intracellular increase in cyclic adenosine monophosphate (cAMP). Higher cAMP levels in the cell induce protein kinase A activation, which in turn leads to phosphorylation of ion channels, resulting in Cl release as well as decrease in Na+ uptake and, consequently, massive water release to intestinal lumen, the major characteristic of secretory diarrhea caused by these pathogens.6,341 ST, a monomeric protein of about 5kDa, may also induce osmotic deregulation, activating directly the guanylate cyclase C located at the apical membrane of the intestinal cells to produce intracellular cyclic guanosine monophosphate and consequently to generate secretion of Cl ions and water from the intestinal epithelium. However, an ST variant first isolated from pigs shows distinct physiological activity, characterized by the loss of villus epithelial cells and net bicarbonate secretion.6 The toxins LT and ST, separately or in combination, are able to induce cellular water-electrolyte imbalance, which surely contributes to ETEC pathogenesis.

A hallmark in ETEC biology is the expression of enterotoxins, which also display a significant antigenic heterogeneity. Approximately one-third of the strains isolated from diarrheic patients express only LT or only ST, while another third express both toxin types. In addition, two unrelated ST groups, with different functional and structural features, have been identified: (i) STa, comprising two variants (STh and STp) associated with human disease, and (ii) STb, which is generally found among swine-derived ETEC strains. Similarly, LT are divided into two antigenically distinct groups: LT-I and LT-II.6 Initially, two LT-I variants, isolated from human or swine-derived ETECs (LTh and LTp, respectively), were described and shown to have high amino acid sequence identity and similar but not equal antigenicity and biochemical and receptor-binding properties.342,343 The related LT-II variants (LT-IIa,-IIb,-IIc) have been isolated from human beings or other hosts and contaminated food and bind to different receptors.344–347 The LT-IIa, LT-IIb and LT-IIc share 51, 52 and 49% or 15, 16 and 7% identity with LT-Ih regarding the A and B subunits, respectively.347,348

More recently, a pioneer study carried out with ETEC strains isolated in Brazil demonstrated a rather high intraspecific LTh variability between LT-producing ETEC strains.333,349,350 In a collection of 51 ETEC strains expressing LT and/or ST, 50 genetic polymorphic sites were found in the LT-encoding genes, which revealed 16 natural LT variants according to differences in amino acid sequences. Among these variants, named LT1 to LT16, two (LT1 and LT2) were associated with a limited number of serotypes with a global distribution and mainly isolated from diarrheic patients.333 In contrast, most of the detected LT variants were observed among LT-producing ETEC strains isolated from asymptomatic subjects.333 More recently, 12 additional LT types were identified in a larger collection of ETEC strains isolated from different regions of the world.351 Interestingly, a much reduced genetic variability was found in the LT-encoding genes among ETEC strains isolated from pigs (LTp) and ST-encoding genes.352,353

The natural diversity of LT types found among ETEC strains isolated from symptomatic and asymptomatic humans suggests that some LT types can show higher toxicity to eukaryotic cells and can be expressed at different levels compared to other toxin types. Indeed, previous observations indicated that some LT types are endowed with different toxicity, under in vitro and in vivo conditions.333,349,350 A natural LT variant, similar to the LT expressed by swine-derived strains, showed reduced toxicity due to an amino acid replacement at a key polymorphic site in the A subunit.333,350 This amino acid change provided a less flexible A subunit structure, impairing appropriate contact with the cofactor (NAD+) at the catalytic site.350 Other authors observed that natural polymorphisms in the B subunit resulted in decreased receptor binding and therefore reduced toxicity to eukaryotic cells.352 These results suggest that the presence of ETEC strains expressing different LT variants may correlate with the incidence of symptoms among infected subjects, particularly among infected infants not previously exposed to ETEC infections.

Variable LT expression may also impact the severity of ETEC-associated disease. Previous observations demonstrated that the amounts of LT produced and/or secreted by ETEC are dramatically different among strains and clinical isolates.332,351,354–356 The presence of single nucleotide changes in the etx operon regulatory region may be found and, at least for some of them, are associated with different transcriptional and translational activity among wild ETEC strains.332,unpublished data Nonetheless, further studies are required to demonstrate a clear link between transcriptional and post-transcriptional events and the severity of the symptoms associated with ETEC infection.

Epidemiology

Annually, infections with different ETEC strains cause an astonishing number of diarrheal episodes, greatly exceeding 200 million cases and causing approximately 75,000 deaths, mainly among babies and young children in tropical areas with poor sanitary conditions.118,357 In Brazil, epidemiological data harvested at different times between 1978 and 2007 have demonstrated that the incidence of ETEC-induced diarrhea ranges from 3.5 to 20.45%.115,358–361

Detection and diagnosis

This pathotype is mainly characterized by the enterotoxins it produces, and diagnosis depends upon identifying either LT and/or ST. One or both toxins may be expressed by ETEC strains.340,362–364 The diagnosis of ETEC strains should include, in addition to LT and ST detection, complementary PCR assays for the detection of virulence genes such as clyA, eatA, tia, tibC, leoA, and east-1.340 A sensitive and specific PCR assay with primers targeting the genes lt and st was reported by Stacy-Phipps et al.,365 and later by Youmans et al.,366 using quantitative real-time PCR. Moreover, several multiplex PCR assays were also developed using these two genes.367–369

Phenotypical detection of ETEC was initially performed using supernatants obtained from single E. coli colonies and by laborious procedures such as rabbit ileal loop test,370 suckling mouse assay371 or cytopathic effect studies on CHO or Y1 adrenal cell monolayers, in which the presence of LT in supernatants was indicated by rounding of Y1 cells or elongation of CHO cells after 24h of incubation.372,373

A number of immunoassays have been developed for ST detection, including radioimmunoassay and enzyme-linked immunosorbent assay (ELISA). Both tests correlate well with results obtained with the suckling-mouse assay and require substantially less expertise.374,375 ELISA assays were then developed using the GM1 receptor to bind LT obtained from filtered culture supernatants or employing a competitive test for LT, which replaced former procedures.376

Immunological assays for LT detection includes the traditional Biken test, latex agglutination, and reliable and easy to perform commercially available tests, such as the reversed passive latex agglutination and the staphylococcal coagglutination test.321 Several immunological assays where LT is captured either by ganglioside GM1 (its receptor in the host cell) or by antibodies have been described.139,321,377,378 Assays for ST by indirect ELISA using IgG1 ST-mAb and for LT by capture ELISA employing IgG enriched fraction of a rabbit polyclonal as a capture antibody and IgG2b LT-mAb as a second antibody have been employed as tools for diagnosis. The presence of bile salts and the use of certain antibiotics improved ETEC toxin production/release. Triton X-100, as chemical treatment, proved to be an alternative method for toxin release. Consequently, a common protocol that can increase the production and release of LT and ST could facilitate and enhance the sensitivity of diagnostic tests for ETEC.355 Afterwards, those monoclonal antibodies were rebuilt resulting in single chain fragment variable (scFv) fragments. The developed recombinant scFvs against LT and ST constitute a promising starting point for simple and cost-effective ETEC diagnosis.379

Enteroinvasive E. coli

Enteroinvasive E. coli (EIEC) is a causative agent of dysentery in humans, especially in developing countries.380 It causes keratoconjunctivitis in experimental guinea pigs381 and invades human colon cells, causing an infection similar to that caused by Shigella sp.382,383 The first description of EIEC was performed by EWING and GRAWATTI in 1947.384 The first works emphasizing the particular biochemical characteristics of EIEC samples were presented in 1967 by Trabulsi et al.,385 in Brazil and by Sakazaki et al.,386 in Japan. All isolates studied were Serény test positive (guinea pig keratoconjunctivitis) and the strains were lysine decarboxylase negative, late fermenting lactose and generally non-motile, except for samples of the O124 serogroup. The study of the biochemical behavior of 97 samples of EIEC381 corroborated the results obtained previously. It has been shown that this group of diarrheagenic E. coli belonged to well-defined bioserotypes, O28ac:H-, O29:H-, O112ac:H-, O121:H-, O124:H-, O124:H30, O135:H-, O136:H-, O143:H, O144:H-, O152:H-, O159:H-, O164:H-, O167:H- and O173:H-.381,387–389 In 1964, it was demonstrated that samples of the O32 and O42 serogroups of E. coli also had the ability to cause keratoconjunctivitis in guinea pigs.389 However, the existence of enteroinvasive bioserotypes in O42 serogroup was not confirmed, and O32 bioserotype is actually an aerogenic variant of S. boydii 14, as shown by Toledo et al.390 There are reports of isolation of EIEC samples belonging to other mobile serotypes, O144H25391; however, these are sporadic cases. Recently, the serotype of E. coli O96:H19 was described as enteroinvasive E. coli in two large outbreaks occurring in Italy and United Kingdom.392,393 It is worth mentioning that EIEC serotypes considered to be nonmotile produce an unusually large (77kDa) flagellin that is assembled into functional flagellum filaments that allow the bacteria to swim in modified motility agar (0.2%).394 Analysis of the fliC gene showed that 11 different EIEC serotypes have six molecular profiles of fliC. The major EIEC serotypes showed low fliC diversity. The dendrogram showed two major clusters, suggesting two different origins for the flagellin gene among these strains. In addition, the presence of the same pattern among strains of the same serotype suggests the existence of a common clone.395

Virulence factors, mechanisms and pathogenesis

Diarrhea due to EIEC and Shigella is caused by the invasion and penetration of bacteria in the enterocytes, leading to their destruction. These bacteria bind specifically to the mucosa of the large intestine and invade cells by endocytosis.396,397Shigella flexneri strains are used as template for most studies of invasion.

The complex process in colonization and EIEC survival in the gastrointestinal barrier depends on the presence of a large plasmid of about 220kb (pInv), very similar to that found in Shigella.397–400 In this process, multiple bacterial genes are involved, both chromosomal and plasmidial. Bacteria without the virulence plasmid do not cause keratoconjunctivitis in guinea pigs, being considered non-virulent.397,401

Most of these functions are related to proteins encoded by a 31-kb fragment from pInv, containing 38 genes. In this fragment are genes responsible for bacterial invasion and escape, by cell spreading, inhibition of autophagy, regulation of immune response of the host apparatus and type III secretion system (TTSS). Once injected into the host cell, the virulence or effector factors induce or inhibit cell signaling pathways. The changes in host cells induced by bacteria allow intracellular survival of these microorganisms.402–404

Due to the great similarity between Shigella and EIEC, it can be assumed that the two would share the same ancestor and that at a given moment in evolution there was a division. Why has EIEC retained some E. coli properties that have been lost in multiple lineages of Shigella? Data obtained by different groups lead to the speculation that EIEC strains are in an intermediate stage and are a potential pre-cursor of “full-blown” Shigella strains.405–409

Despite the similarities invasion mechanism and symptoms of the disease (dysentery), the infectious dose of EIEC is much higher than that of Shigella.410 Furthermore, the disease caused by EIEC appears to be a milder and self-limiting form.

In the Serény test, it was observed that EIEC induces a milder form of the disease (mild/moderate inflammation), while Shigella leads to an exacerbation of proinflammatory response (severe inflammation). Furthermore, keratoconjunctivitis develops more rapidly in guinea pigs inoculated with Shigella (two days) than in guinea pigs inoculated with EIEC (4–5 days).411

Samples from different serotypes of EIEC have shown polymorphism in some regions of genes involved in invasion. However, the data reveal that there are no changes in genes of the invasion plasmid antigens that could explain the differences in pathogenicity between Shigella and EIEC.400 Moreover, recent studies from our group showed that the genes responsible for cell spreading (icsA and icsB) and regulation of the immune response of the host (osp) did not indicate changes that could explain the difference in pathogenicity between Shigella and EIEC (data not shown).

Another important aspect of bacterial colonization is the uptake of iron (Fe) under conditions limited in the host. Iron is an essential element for all living organisms, it is estimated that the microorganisms require iron at concentrations from 10 to 10−6 M to meet their metabolic needs. It was shown that EIEC has a high adaptability, using, if necessary, the iron capture system that consumes less energy. The ability to capture Fe from different sources can facilitate the development of infectious processes by this bacterium.412,413

EIEC, like other enteric pathogens, target M cells (microfold cells) present in the intestinal mucosa as a route of entry to deeper tissues of the host.403,414 Reaching the lamina through the M cells, the bacterial cells are phagocytized by macrophages and dendritic cells. These cells are the first step in the production of the inflammatory response against bacterial invasion. After escape from macrophages and dendritic cells, EIEC are able to invade enterocyte cells from the basolateral side, escaping from the phagosome and replicating in the cytoplasm.403,414

Our group first described the phenotypic and genotypic characteristics explaining the lower capacity of EIEC to cause disease when compared with the species of Shigella. To this end, use has been made of experimental models that mimick the intestinal microenvironment of the host, such as cultures of intestinal epithelial cells, macrophages and dendritic cells.411,415,416 Our results showed that the initial ability to invade the intestinal cell is similar between EIEC and Shigella, but that the expression of virulence genes (ipaABCD, icsA, icsB, virF, virB), capacity to escape from the phagosome, intracellular proliferation and dissemination of EIEC, as well as the ability to cause cell damage during the infection, are much lower than with S. flexneri.411 A significantly greater number of EIEC are seen inside macrophages compared to Shigella after phagocytosis. Furthermore, Shigella shows greater capacity to escape from macrophages as compared to EIEC. The expression of virulence genes, production of proinflammatory cytokines and cell death was found to be less in macrophages infected by EIEC when compared to Shigella. It should be noted that the production of antiinflammatory cytokine IL-10 by macrophages is greater in infection by EIEC than Shigella.415

EIEC interaction with dendritic cells has been evaluated. The data suggest that EIEC induces the production of IL-10, IL-12 and TNF-α by infected dendritic cells, while S. flexneri induce TNF-α production. Unlike Shigella, infection with EIEC increases the expression of TLR-4 and TLR-5 receptors on dendritic cells and decreases the expression of costimulatory molecules that may cooperate to induce the proliferation of T-lymphocytes, and in addition, there is a greater proliferation of lymphocytes challenged with S. flexneri than with EIEC.416

Epidemiology

The EIEC strains have similar biochemical, genetic and pathogenic characteristics as Shigella species, which can often make the correct identification of this pathotype difficult.381,385,386,417 Epidemiological data may be underestimated due to the difficulty in differentiating between Shigella and EIEC.

EIEC was responsible for several outbreaks, but there are few reports on routes of transmission and distribution of this bacterium in nature. Water and cheese were described as potential sources,418–421 as well as the direct transmission through person-to-person contact.422 In the 1970s, a major outbreak of diarrhea was reported in the United States, which affected 387 patients. The transmission vehicle was an imported cheese, contaminated by O124 serogroup.419 According to the food and drug control agency of the United States (Food and Drug Administration-FDA), outbreaks caused by EIEC have been associated with milk and milk products and beef; however, any food or water contaminated with human feces of an individual patient can cause disease in other individuals.423 In Brazil, there is a report of three samples isolated from water.421 Outbreaks involving two EIEC were recently reported in Europe, one in Italy in 2012 involving 109 cases and another in the United Kingdom in 2014 involving 50 cases.392,393 In both, vegetables were to blame.

In Calcutta, the prevalence of EIEC in a group of 263 patients hospitalized with diarrhea was high, 16.3% of cases.424 However, there are reports of a prevalence of 2%.425 In Thailand, China and other Asian countries, a prevalence of 4 to 7% has been seen.426–429 In Bolivia, the reports showed a 2% prevalence.430 Some studies have shown that in Nigeria, Iran and Thailand, the distribution of EIEC is below (less than 0.1%) the rates found in developed countries; in Spain, for example, a prevalence of 0.2% was found.431–434 The low incidence can be due to difficulties in differentiating EIEC from Shigella.

The isolation of EIEC in Brazil has ranged from 0.5 to 15%, depending on the population investigated.435–440 The data suggest that the presence of EIEC is related to socioeconomic conditions. Toledo and Trabulsi439 investigated the presence of this microorganism from children under five years of age and non-slum-dwelling children from different areas of the city of São Paulo. This bacterium has been found in 17 of 107 slum-dwelling children with diarrhea (15.9%) and in 16 of 701 non-slum-dwelling children with diarrhea (2.3%). In the first group, EIEC was the enteropathogen most frequently isolated from children over 2 years of age. In non-slum-dwelling children of the same age, it was the fourth most common agent, being more frequent than EPEC, Salmonella, Rotavirus, and Yersinia enterocolitica. Studies performed outside the city of São Paulo showed a low prevalence of these bacteria, 0.5–2.5%.435,440

Detection and diagnosis

Samples of EIEC grow well in culture medium routinely used for isolation of Enterobacteriaceae, such as MacConkey agar, xylose-lysine-deoxycholate (XLD) agar and Hektoen enteric (HE). Highly selective media such as Salmonella Shigella agar (SS) or bismuth sulfite agar may not be as effective for some serotypes.381

The identification of E. coli species may be carried out using conventional biochemical tests, such as production of indole, fermentation of glucose, sucrose and lactose, gas production from glucose fermentation, pathway glucose fermentation, using citrate as sole carbon source, motility, lysine, arginine and ornithine decarboxylation.1,441 Fermentation of lactose varies according to the strain; EIEC samples can ferment lactose slowly (72h), making it difficult to differentiate from Shigella.381 Complementary to the physiological and biochemical characteristics, serotyping may be required for the differentiation, since some serotypes of S. flexneri produce indole. In such cases, O antisera of EIEC and Shigella should be used.1,441 Bacterial colonies with this characteristic can be screened for the classical EIEC serogroups O28ac, O29, O112, O124, O136, O143, O144, O152, O159, O164, O169, and O173.1,441,443 EIEC invasive capacity can be evaluated using the Sereny guinea pig eye test444 and tissue culture assays,445 which are more markedly limited to reference laboratories.

To characterize the EIEC pathotype, it is necessary to search for plasmid virulence genes. Currently, the investigation of the ipaH gene, a multi-copy gene (4–10) present in EIEC and Shigella, by PCR is recommended,442,446 or studies of other DNA sequences are needed, such as the invasion-associated locus gene (ial).447 The presence of the iudA and lacY genes can differentiate EIEC from S. flexneri.446 A simple and rapid stool test based on apyrase (ATP-diphosphohydrolase) activity was described for EIEC detection.448 This is an essential periplasmic enzyme required for unipolar localization of IcsA, which is involved in the pathogen's intracellular and intercellular spread, and is only expressed by EIEC and Shigella.449 The enzyme activity is measured by a colorimetric reaction. The method is robust, requires widely available equipment and affordable reagents, and can be applied for routine use in laboratories with limited resources.448

Conclusions

The genomic plasticity of E. coli strains is noteworthy, as can be seen by the variety of strains ranging from commensal residents of the gastrointestinal tract to assorted pathogens that are able to promote intestinal or extraintestinal illnesses with different clinical consequences. It is thus important to note that the continuous evolution of the E. coli genome has hindered the classification of certain E. coli isolates into a pathotype, because some isolates combine the main virulence characteristics of different pathotypes and are thus considered hybrid pathotypes (reviewed in 5) with the potential of allowing the rise of new and more virulent pathogenic E. coli hybrids.

Whole-genome sequencing has provided a great amount of useful information on the genome of pathogenic E. coli, which will help improve diagnosis, typing, disease management, epidemiology and outbreak investigations as well as helping to monitor the spread of pathogens.5 Despite the recent advances in our knowledge of the genetic background and pathogenicity of strains of different DEC pathotypes, various novel genes encoding unknown functions are yet to be characterized to further our understanding of the interactions of these pathogens with their hosts.

Conflicts of interest

The authors declare no conflicts of interest.

Acknowledgement

The authors dedicate the present article to Prof. Luiz R. Trabulsi who, during his life, inspired us and several others in the study of microbiology, particularly in E. coli pathogenesis. He also left to all who met him in life a wonderful and unforgettable example of professional dedication and ethical commitment in science and education.

References
[1]
W.H. Ewing.
Edwards and Ewing's Identification of Enterobacteriaceae.
4th ed., Elsevier, (1986),
[2]
P.L. Conway.
Microbial ecology of the human large intestine.
Human Colonic Bacteria: Role in Nutrition, Physiology and Pathology, pp. 1-24
[3]
J.B. Kaper, J.P. Nataro, H.L.T. Mobley.
Pathogenic Escherichia coli.
Nat Rev Microbiol, 2 (2004), pp. 123-140
[4]
World Health Organization.
World Health Statistics.
WHO Press, (2012),
[5]
M.A. Croxen, R.J. Law, R. Scholz, K.M. Keeney, M. Wlodarska, B.B. Finlay.
Recent advances in understanding enteric pathogenic Escherichia coli.
Clin Microbiol Rev, 26 (2013), pp. 822-880
[6]
J.P. Nataro, J.B. Kaper.
Diarrheagenic Escherichia coli.
Clin Microbiol Rev, 11 (1998), pp. 142-201
[7]
N. Rolhion, A. Darfeuille-Michaud.
Adherent-invasive Escherichia coli in inflammatory bowel disease.
Inflamm Bowel Dis, 13 (2007), pp. 1277-1283
[8]
R.J. Cieza, A.T. Cao, Y. Cong, A.G. Torres.
Immunomodulation for gastrointestinal infections.
Expert Rev Anti Infect Ther, 10 (2012), pp. 391-400
[9]
E. Neter, O. Westphal, O. Luderitz, R.M. Gino, E.A. Gorzynski.
Demonstration of antibodies against enteropathogenic Escherichia coli in sera of children of various ages.
Pediatrics, 16 (1995), pp. 801-807
[10]
J. Bray.
Isolation of antigenically homogeneous strains of Bacterium coli neopolitanum from summer diarrhoea of infants.
J Pathol Bacteriol, 57 (1945), pp. 239-247
[11]
R.M. Robins-Browne.
Traditional enteropathogenic Escherichia coli of infantile diarrhea.
Rev Infect Dis, 9 (1987), pp. 28-53
[12]
L.R. Trabulsi, R. Keller, T.A.T. Gomes.
Typical and atypical enteropathogenic Escherichia coli.
Emerg Infect Dis, 8 (2002), pp. 508-513
[13]
F. Ørskov, T.S. Whittam, A. Cravioto, I. Ørskov.
Clonal relationships among classic enteropathogenic Escherichia coli (EPEC) belong to different O groups.
J Infect Dis, 162 (1990), pp. 76-81
[14]
T.S. Whittam, E.A. McGraw.
Clonal analysis of EPEC serogroups.
Rev Microbiol, 27 (1996), pp. 7-16
[15]
T.A.T. Gomes, B. González-Pedrajo.
Enteropathogenic Escherichia coli (EPEC).
Pathogenic Escherichia coli in Latin America, pp. 66-126
[16]
T.A.T. Gomes, P.M. Griffin, C. Ivey, L.R. Trabulsi, S.R.T.S. Ramos.
EPEC infections in São Paulo.
International Symposium on Enteropathogenic Escherichia coli (EPEC), São Paulo, SP, 27 (1996), pp. 25-33
[17]
T.H. Hazen, J.W. Sahl, C.M. Fraser, M.S. Donnenberg, F. Scheutz, D.A. Rasko.
Refining the pathovar paradigm via phylogenomics of the attaching and effacing Escherichia coli.
PNAS, 110 (2013), pp. 12810-12815
[18]
R.T. Hernandes, W.P. Elias, A.M. Vieira, T.A.T. Gomes.
An overview of atypical enteropathogenic Escherichia coli.
FEMS Microbiol Lett, 297 (2009), pp. 137-149
[19]
M.M. Levine, J.P. Nataro, H. Karch, et al.
The diarrheal response of humans to some classic serotypes of enteropathogenic Escherichia coli is dependent on a plasmid encoding an enteroadhesiveness factor.
J Infect Dis, 152 (1985), pp. 550-559
[20]
M.A. Vieira, J.R. Andrade, L.R. Trabulsi, et al.
Phenotypic and genotypic characteristics of Escherichia coli strains of non-enteropathogenic E. coli (EPEC) serogroups that carry eae and lack the EPEC adherence factor and Shiga toxin DNA probe sequences.
J Infect Dis, 183 (2001), pp. 762-772
[21]
I.C. Scaletsky, M.L. Silva, L.R. Trabulsi.
Distinctive patterns of adherence of enteropathogenic Escherichia coli to HeLa cells.
Infect Immun, 45 (1984), pp. 534-536
[22]
R. Rothbaum, A.J. McAdams, R. Giannella, J.C. Partin.
A clinicopathological study of enterocyte-adherent Escherichia coli: a cause of protracted diarrhea in infants.
Gastroenterology, 83 (1982), pp. 441-454
[23]
J.A. Girón, A.S. Ho, G.K. Schoolnik.
An inducible bundle-forming pilus of enteropathogenic Escherichia coli.
Science, 254 (1991), pp. 710-713
[24]
D. Bieber, S.W. Ramer, C.Y. Wu, et al.
Type IV pili, transient bacterial aggregates, and virulence of enteropathogenic Escherichia coli.
Science, 280 (1998), pp. 2114-2118
[25]
J. Vuopio-Varkila, G.K. Schoolnik.
Localized adherence by enteropathogenic Escherichia coli is an inducible phenotype associated with the expression of new outer membrane proteins.
J Exp Med, 174 (1991), pp. 1167-1177
[26]
C.G. Moreira, K. Palmer, M. Whiteley, et al.
Bundle-forming pili and EspA are involved in biofilm formation by enteropathogenic Escherichia coli.
J Bacteriol, 188 (2006), pp. 3952-3961
[27]
R.M. Hyland, J. Sun, T.P. Griener, et al.
The bundling pilin protein of enteropathogenic Escherichia coli is an N-acetyllactosamine-specific lectin.
Cell Microbiol, 10 (2008), pp. 177-187
[28]
K.D. Stone, H. Zhang, L.K. Carlson, M.S. Donnenberg.
A cluster of fourteen genes from enteropathogenic Escherichia coli is sufficient for the biogenesis of a type IV pilus.
Mol Microbiol, 20 (1996), pp. 325-337
[29]
T. Tobe, T. Hayashi, C. Han, G.K. Schoolnik, E. Ohtsubo, C. Sasakawa.
Complete DNA sequence and structural analysis of the enteropathogenic Escherichia coli adherence factor plasmid.
Infect Immun, 67 (1999), pp. 5455-5462
[30]
C. Brinkley, V. Burland, R. Keller, et al.
Nucleotide sequence analysis of the enteropathogenic Escherichia coli adherence factor plasmid pMAR7.
Infect Immun, 74 (2006), pp. 5408-5413
[31]
T.H. Hazen, J.B. Kaper, J.P. Nataro, D.A. Rasko.
Comparative genomics provides insight into the diversity of the attaching and effacing Escherichia coli virulence plasmids.
Infect Immun, 83 (2015), pp. 4103-4117
[32]
J.P. Nougayrède, P.J. Fernandes, M.S. Donnenberg.
Adhesion of enteropathogenic Escherichia coli to host cells.
Cell Microb, 5 (2003), pp. 359-372
[33]
H.W. Moon, S.C. Whipp, R.A. Argenzio, M.M. Levine, R.A. Giannella.
Attaching and effacing activities of rabbit and human enteropathogenic Escherichia coli in pig and rabbit intestines.
Infect Immun, 41 (1983), pp. 1340-1351
[34]
T.K. McDaniel, K.G. Jarvis, M.S. Donnenberg, J.B. Kaper.
A genetic locus of enterocyte effacement conserved among diverse enterobacterial pathogens.
Proc Natl Acad Sci USA, 92 (1995), pp. 1664-1668
[35]
S.J. Elliott, V. Sperandio, J.A. Giron, et al.
The locus of enterocyte effacement (LEE)-encoded regulator controls expression of both LEE- and non-LEE-encoded virulence factors in enteropathogenic Escherichia coli.
Infect Immun, 68 (2000), pp. 6115-6126
[36]
W. Deng, J.L. Puente, S. Grunheid, et al.
Dissecting virulence: systematic and functional analyses of a pathogenicity island.
Proc Natl Acad Sci USA, 101 (2004), pp. 3597-3602
[37]
P. Dean, B. Kenny.
The effector repertoire of enteropathogenic E. coli ganging up on the host cell.
Curr Opin Microbiol, 12 (2009), pp. 101-109
[38]
J. Barba, V.H. Bustamante, M.A. Flores-Valdez, W. Deng, B.B. Finlay, J.L. Puente.
A positive regulatory loop controls expression of the locus of enterocyte effacement-encoded regulators Ler and GrlA.
J Bacteriol, 187 (2005), pp. 7918-7930
[39]
B. Kenny, R. DeVinney, M. Stein, D.J. Reinscheid, E.A. Frey, B.B. Finlay.
Enteropathogenic Escherichia coli (EPEC) transfers its receptor for intimate adherence into mammalian cells.
Cell, 91 (1997), pp. 511-520
[40]
G. Frankel, D.C.A. Candy, P. Everest, G. Dougan.
Characterization of the C-terminal domains of intimin-like proteins of enteropathogenic and enterohemorrhagic Escherichia coli, Citrobacter freundii, and Hafnia alvei.
Infect Immun, 62 (1994), pp. 1835-1842
[41]
D.W. Lacher, H. Steinsland, T.S. Whittam.
Allellic subtyping of the intimin locus (eae) of pathogenic Escherichia coli by fluorescent RFLP.
FEMS Microbiol Lett, 261 (2006), pp. 80-87
[42]
D.W. Lacher, H. Steinsland, T.E. Blank, M.S. Donnenberg, T.S. Whittam.
Molecular evolution of typical enteropathogenic Escherichia coli: clonal analysis by multilocus sequence typing and virulence gene allelic profiling.
J Bacteriol, 189 (2007), pp. 342-350
[43]
M.-H. Ruchaud-Sparagano, S. Muhlen, P. Dean, B. Kenny.
The enteropathogenic Escherichia coli (EPEC) Tir effector inhibits NF-kB activity by targeting TNFα receptor-associated factors.
PLoS Pathog, 7 (2011), pp. e1002414
[44]
A. Santos, B.B. Finlay.
Bringing down the host: enteropathogenic and enterohaemorrhagic Escherichia coli effector-mediated subversion of host innate immune pathways.
Cell Microbiol, 17 (2015), pp. 318-332
[45]
A.R.C. Wong, J.S. Pearson, M.D. Bright, et al.
Enteropathogenic and enterohaemorrhagic Escherichia coli: even more subversive elements.
Mol Microbiol, 80 (2011), pp. 1420-1438
[46]
A. Vossenkämper, T.T. MacDonald, O. Marchès.
Always one step ahead: how pathogenic bacteria use the type III secretion system to manipulate the intestinal mucosal immune system.
J Inflamm, 8 (2011), pp. 11
[47]
B. Raymond, J.C. Young, M. Pallett, R.G. Endres, A. Clements, G. Frankel.
Subversion of trafficking, apoptosis, and innate immunity by type III secretion system effectors.
Trends Microbiol, 21 (2013), pp. 430-441
[48]
J.N. Fletcher, H.E. Embaye, B. Getty, R.M. Batt, C.A. Hart, J.R. Saunders.
Novel invasion determinant of enteropathogenic Escherichia coli plasmid pLV501 encodes the ability to invade intestinal epithelial cells and HEp-2 cells.
Infect Immun, 60 (1992), pp. 2229-2236
[49]
I.C.A. Scaletsky, M.S.V. Gatti, F.J. Da Silveira, I.M. DeLuca, E. Freymuller, L.R. Travassos.
Plasmid encoding for drug resistance and invasion of epithelial cells in enteropathogenic Escherichia coli O111:H-.
Microb Pathog, 18 (1995), pp. 387-399
[50]
J.M. Klapproth, I.C.A. Scaletsky, B.P. McNamara, et al.
A large toxin from pathogenic Escherichia coli strains that inhibits lymphocyte activation.
Infect Immun, 68 (2000), pp. 2148-2155
[51]
L. Nicholls, T.H. Grant, R.M. Robins-Browne.
Identification of a novel genetic locus that is required for in vitro adhesion of a clinical isolate of enterohaemorrhagic Escherichia coli to epithelial cells.
Mol Microbiol, 35 (2000), pp. 275-288
[52]
I. Tatsuno, M. Horie, H. Abe, et al.
toxB gene on pO157 of enterohemorrhagic Escherichia coli O157:H7 is required for full epithelial cell adherence phenotype.
Infect Immun, 69 (2001), pp. 6660-6669
[53]
L. Badea, S. Doughty, L. Nicholls, J. Sloan, R.M. Robins-Browne, E.L. Hartland.
Contribution of Efa/LifA to the adherence of enteropathogenic Escherichia coli to epithelial cells.
Microb Pathog, 34 (2003), pp. 205-215
[54]
J.M. Klapproth, M. Sasaki, M. Sherman, et al.
Citrobacter rodentium lifA/efa1 is essential for colonic colonization and crypt cell hyperplasia in vivo.
Infect Immun, 73 (2005), pp. 1441-1451
[55]
I. Tatsuno, R. Mundy, G. Frankel, et al.
The lpf gene cluster for long polar fimbriae is not involved in adherence of enteropathogenic Escherichia coli or virulence of Citrobacter rodentium.
Infect Immun, 74 (2006), pp. 265-272
[56]
A.G. Torres, K.J. Kanack, C.B. Tutt, V. Popov, J.B. Kaper.
Characterization of the second long polar (LP) fimbriae of Escherichia coli O157:H7 and distribution of LP fimbriae in other pathogenic E. coli strains.
FEMS Microbiol Lett, 238 (2004), pp. 333-344
[57]
Z. Saldaña, A.L. Erdem, S. Schuller, et al.
The Escherichia coli common pilus and the bundle-forming pilus act in concern during the formation of localized adherence by enteropathogenic E. coli.
J Bacteriol, 191 (2009), pp. 3451-3461
[58]
D.M. Girão, V.B.C. Girão, K. Irino, T.A.T. Gomes.
Classifying Escherichia coli.
Emerg Infect Dis, 12 (2006), pp. 1297-1298
[59]
B.G. Garcia, T. Ooka, Y. Gotoh, et al.
Genetic relatedness and virulence properties of enteropathogenic Escherichia coli strains of serotype O119:H6 expressing localized adherence or localized and aggregative adherence-like patterns on HeLa cells.
Int J Med Microbiol, 306 (2016), pp. 152-164
[60]
J.A. Girón, A.G. Torres, E. Freer, J.B. Kaper.
The flagella of enteropathogenic Escherichia coli mediate adherence to epithelial cells.
Mol Microbiol, 44 (2002), pp. 361-479
[61]
J. Cleary, L.-C. Lai, R.K. Shaw, et al.
Enteropathogenic Escherichia coli (EPEC) adhesion to intestinal epithelial cells: role of bundle-forming pili (BFP), EspA filaments and intimin.
Microbiology, 150 (2004), pp. 527-538
[62]
T. Yamamoto, N. Wakisaka, F. Sato, A. Kato.
Comparison of the nucleotide sequence of enteroaggregative Escherichia coli heat-stable enterotoxin 1 genes among diarrhea-associated Escherichia coli.
FEMS Microbiol Lett, 147 (1997), pp. 89-96
[63]
M.V. Dulguer, S.H. Fabricotti, S.Y. Bando, C.A. Moreira-Filho, U. Fagundes-Neto, I.C.A. Scaletsky.
Atypical enteropathogenic Escherichia coli strains: phenotypic and genetic profiling reveals a strong association between enteroaggregative E. coli heat-stable enterotoxin and diarrhea.
J Infect Dis, 188 (2003), pp. 1685-1694
[64]
L.E.P. Silva, T.B. Souza, N.P. Silva, I.C.A. Scaletsky.
Detection and genetic analysis of the enteroaggregative Escherichia coli heat-stable enterotoxin (EAST1) gene in clinical isolates of enteropathogenic Escherichia coli (EPEC) strains.
BMC Microbiol, 14 (2014), pp. 135
[65]
A.C.R. Ghilardi, T.A.T. Gomes, L.R. Trabulsi.
Production of Cytolethal Distending Toxin and other virulence characteristics of Escherichia coli strains of serogroup O86.
Mem Inst Oswaldo Cruz, 96 (2001), pp. 703-708
[66]
I.R. Henderson, F. Navarro-Garcia, M. Desvaux, R.C. Fernandez, D. Ala’Aldeen.
Type V protein secretion pathway: the autotransporter story.
Microbiol Mol Biol Rev, 68 (2004), pp. 692-744
[67]
T.A. Gomes, D. Yamamoto, M.A.M. Vieira, R.T. Hernandes.
Atypical enteropathogenic Escherichia coli.
Escherichia coli in the Americas, pp. 77-96
[68]
J.E. Vidal, F. Navarro-Garcia.
EspC translocation into epithelial cells by enteropathogenic Escherichia coli requires a concerted participation of type V and III systems.
Cell Microbiol, 10 (2008), pp. 1976-1986
[69]
M.E. Drago-Serrano, S.G. Parra, H.A. Manjarrez-Hernández.
EspC, an autotransporter protein secreted by enteropathogenic Escherichia coli (EPEC), displays protease activity on human hemoglobin.
FEMS Microbiol Lett, 265 (2006), pp. 35-40
[70]
N. Salinger, B. Kokona, R. Fairman, I. Okeke.
The plasmid-encoded regulator activates factors conferring lysozyme resistance on enteropathogenic Escherichia coli strains.
Appl Environ Microbiol, 75 (2009), pp. 275-280
[71]
F. Navarro-Garcia, A. Serapio-Palacios, J.E. Vidal, M.I. Salazar, G. Tapia-Pastrana.
EspC promotes epithelial cell detachment by enteropathogenic Escherichia coli via sequential cleavages of a cytoskeletal protein and then focal adhesion proteins.
Infect Immun, 82 (2014), pp. 2255-2265
[72]
J. Xicohtencatl-Cortes, Z. Saldaña, W. Deng, et al.
Bacterial macroscopic ropelike fibers with cytopathic and adhesive properties.
J Biol Chem, 285 (2010), pp. 32336-32342
[73]
M.S. Donnenberg, J.B. Kaper.
Enteropathogenic Escherichia coli.
Infect Immun, 60 (1992), pp. 3953-3961
[74]
K.G. Campellone, A. Giese, D.J. Tipper, J.M. Leong.
A tyrosine-phosphorylated 12-aminoacid sequence of enteropathogenic Escherichia coli Tir binds the host adaptor protein Nck and is required for Nck localization to actin pedestals.
Mol Microbiol, 43 (2002), pp. 1227-1241
[75]
M. Bortoloni, L. Trabulsi, R. Keller, G. Frankel, V. Sperandio.
Lack of expression of bundle-forming pili in some clinical isolates of enteropathogenic Escherichia coli (EPEC) is due to a conserved large deletion in the bfp operon.
FEMS Microbiol Lett, 179 (1999), pp. 169-174
[76]
J. Rodrigues, I.C. Scaletsky, L.C. Campos, T.A. Gomes, T.S. Whittam, L.R. Trabulsi.
Clonal structure and virulence factors in strains of Escherichia coli of the classic serogroup O55.
Infect Immun, 64 (1996), pp. 2680-2686
[77]
I.C.A. Scaletsky, J.S. Pelayo, R. Giraldi, J. Rodrigues, M.Z. Pedroso, L.R. Trabulsi.
EPEC adherence to HEp-2 cells.
Rev Microbiol, 27 (1996), pp. 58-62
[78]
C.M. Abe, L.R. Trabulsi, J. Blanco, et al.
Virulence features of atypical enteropathogenic Escherichia coli identified by the eae+ EAF-negative stx-genetic profile.
Diagn Microbiol Infect Dis, 64 (2009), pp. 357-365
[79]
I.C.A. Scaletsky, K.R. Aranda, T.B. Souza, N.P. Silva, M.B. Morais.
Evidence of pathogenic subgroups among atypical enteropathogenic Escherichia coli strains.
J Clin Microbiol, 47 (2009), pp. 3756-3759
[80]
T.A.T. Gomes, R.T. Hernandes, A.G. Torres, et al.
Adhesin-encoding genes from Shiga toxin-producing Escherichia coli are more prevalent in atypical than in typical enteropathogenic E. coli.
J Clin Microbiol, 49 (2011), pp. 3334-3337
[81]
V. Bueris, J. Huerta-Cantillo, F. Navarro-Garcia, R.M. Ruiz, A.M. Cianciarullo, W.P. Elias.
Late establishment of the attaching and effacing lesion caused by atypical enteropathogenic Escherichia coli depends on protein expression regulated by Per.
Infect Immun, 83 (2015), pp. 379-388
[82]
T.A.T. Gomes, K. Irino, D.M. Girão, et al.
Emerging enteropathogenic Escherichia coli strains?.
Emerg Infect Dis, 10 (2004), pp. 1851-1855
[83]
C.A. Contreras, T.J. Ochoa, D.W. Lacher, et al.
Allelic variability of critical virulence genes (eae, bfpA and perA) in typical and atypical enteropathogenic Escherichia coli in Peruvian children.
J Med Microbiol, 59 (2010), pp. 25-31
[84]
Y. Xu, X. Bai, A. Zhao, et al.
Genetic diversity of intimin gene of atypical enteropathogenic Escherichia coli isolated from human, animals and raw meats in China.
PLoS ONE, 11 (2016), pp. e0152571
[85]
M.A. Vieira, L.F. Santos, R.C.B. Dias, et al.
Atypical enteropathogenic Escherichia coli as etiologic agents of sporadic and outbreak-associated diarrhea in Brazil.
J Med Microbiol, 65 (2016), pp. 998-1006
[86]
S.M. Tennant, M. Tauschek, K. Azzopardi, et al.
Characterisation of atypical enteropathogenic E. coli strains of clinical origin.
BMC Microbiol, 9 (2009), pp. 117
[87]
I.C.A. Scaletsky, K.R.S. Aranda, T.B. Souza, N.P. Silva.
Adherence factors in atypical enteropathogenic Escherichia coli strains expressing the localized adherence-like pattern in HEp-2 cells.
J Clin Microbiol, 48 (2010), pp. 302-306
[88]
R.T. Hernandes, I. Velsko, S.C.F. Sampaio, et al.
Fimbrial adhesins produced by atypical enteropathogenic Escherichia coli strains.
Appl Environ Microbiol, 77 (2011), pp. 8391-8399
[89]
I.C. Scaletsky, J. Michalski, A.G. Torres, M.V. Dulguer, J.B. Kaper.
Identification and characterization of the locus for diffuse adherence, which encodes a novel afimbrial adhesin found in atypical enteropathogenic Escherichia coli.
Infect Immun, 73 (2005), pp. 4753-4765
[90]
R.T. Hernandes, A. Miguel, D. Yamamato, et al.
Dissection of the role of pili and Type 2 and 3 secretion systems in adherence and biofilm formation of an atypical enteropathogenic Escherichia coli strain.
Infect Immun, 81 (2013), pp. 3793-3802
[91]
J. Hu, A.G. Torres.
Enteropathogenic Escherichia coli: foe or innocent bystander?.
Clin Microbiol Infect, 21 (2015), pp. 729-734
[92]
S.C.F. Sampaio, W.B. Luiz, M.A.M. Vieira, et al.
Flagellar cap protein FliD mediates adherence of atypical enteropathogenic Escherichia coli to enterocyte microvilli.
Infect Immun, 84 (2016), pp. 1112-1122
[93]
C.T.P. Moraes, J.M. Polatto, S.S. Rossato, et al.
Flagellin and GroEL mediates in vitro binding of an atypical enteropathogenic Escherichia coli to cellular fibronectin.
BMC Microbiol, 15 (2015), pp. 278
[94]
H.F. Culler, C.M. Mota, C.M. Abe, W.P. Elias, M.P. Sircili, M.R. Franzolin.
Atypical enteropathogenic Escherichia coli strains form biofilm on abiotic surfaces regardless of their adherence pattern on cultured epithelial cells.
Biomed Res Int, 2014 (2014), pp. 845147
[95]
H.H. Nascimento, L.E.P. Silva, R.T. Souza, N.P. Silva, I.C.A. Scaletsky.
Phenotypic and genotypic characteristics associated with biofilm formation in clinical isolates of atypical enteropathogenic Escherichia coli (aEPEC) strains.
BMC Microbiol, 14 (2014), pp. 184
[96]
M. Weiss-Muszkat, D. Shakh, Y. Zhou, et al.
Biofilm formation by and multicellular behavior of Escherichia coli O55:H7, an atypical enteropathogenic strain.
Appl Environ Microbiol, 76 (2010), pp. 1545-1554
[97]
J.F. Gärtner, M.A. Schmidt.
Comparative analysis of locus of enterocyte effacement pathogenicity islands of atypical enteropathogenic Escherichia coli.
Infect Immun, 72 (2004), pp. 6722-6728
[98]
D.J. Ingle, M. Tauschek, D.J. Edwards, et al.
Evolution of atypical enteropathogenic E. coli by repeated acquisition of LEE pathogenicity island variants.
Nat Microbiol, 1 (2016), pp. 15010
[99]
D. Müller, I. Benz, A. Liebchen, I. Gallitz, H. Karch, M.A. Schmidt.
Comparative analysis of the locus of enterocyte effacement and its flanking regions.
Infect Immun, 77 (2009), pp. 3501-3513
[100]
T. Ooka, M.A. Vieira, Y. Ogura, et al.
Characterization of tccP2 carried by atypical enteropathogenic Escherichia coli.
FEMS Microbiol Lett, 271 (2007), pp. 126-135
[101]
S.P.D. Rocha, C.M. Abe, V. Sperandio, S.Y. Bando, W.P. Elias.
Atypical enteropathogenic Escherichia coli that contains functional locus of enterocyte effacement genes can be attaching-and-effacing negative in cultured epithelial cells.
Infect Immun, 79 (2011), pp. 1833-1841
[102]
R. Bulgin, A. Arbeloa, D. Goulding, et al.
The T3SS effector EspT defines a new category of invasive enteropathogenic E. coli (EPEC) which form intracellular actin pedestals.
PLoS Pathog, 5 (2009), pp. e1000683
[103]
C. Buss, D. Müller, C. Rüter, G. Heusipp, M.A. Schmidt.
Identification and characterization of Ibe, a novel type III effector protein of A/E pathogens targeting human IQGAP1.
Cell Microbiol, 11 (2009), pp. 661-677
[104]
A. Arbeloa, M. Blanco, F.C. Moreira, et al.
Distribution of espM and espT among enteropathogenic and enterohaemorrhagic Escherichia coli.
J Med Microbiol, 58 (2009), pp. 988-995
[105]
R.T. Hernandes, R.M. Silva, S.M. Carneiro, et al.
The localized adherence pattern of an atypical enteropathogenic Escherichia coli is mediated by intimin omicron and unexpectedly promotes HeLa cell invasion.
Cell Microbiol, 10 (2008), pp. 415-425
[106]
V.C.R. Pacheco, D. Yamamoto, C.M. Abe, et al.
Invasion of differentiated intestinal Caco-2 cells is a sporadic property among atypical enteropathogenic Escherichia coli strains carrying common intimin subtypes.
Pathog Dis, 70 (2014), pp. 167-175
[107]
D. Yamamoto, R.T. Hernandes, M. Blanco, et al.
Invasiveness as a putative additional virulence mechanism of some atypical enteropathogenic Escherichia coli strains with different uncommon intimin types.
BMC Microbiol, 9 (2009), pp. 146
[108]
M.A.M. Vieira, F.A. Salvador, R.M. Silva, et al.
Prevalence and characteristics of the O122 pathogenicity island in typical and atypical enteropathogenic Escherichia coli strains.
J Clin Microbiol, 48 (2010), pp. 1452-1455
[109]
A.G. Abreu, V. Bueris, T.M. Porangaba, M.P. Sircili, F. Navarro-Garcia, W.P. Elias.
Autotransporter protein-encoding genes of diarrheagenic Escherichia coli are found in both typical and atypical enteropathogenic E. coli strains.
Appl Environ Microbiol, 79 (2013), pp. 411-414
[110]
A.G. Abreu, C.M. Abe, K.O. Nunes, et al.
The serine protease Pic as a virulence factor of atypical enteropathogenic Escherichia coli.
Gut Microbes, 7 (2016), pp. 115-125
[111]
R.C. Ruiz, K.C.M. Melo, S.S. Rossato, et al.
Atypical enteropathogenic Escherichia coli secretes plasmid encoded toxin.
Biomed Res Int, 2014 (2014), pp. 896235
[112]
T.J. Ochoa, F. Barletta, C. Contreras, E. Mercado.
New insights into the epidemiology of enteropathogenic Escherichia coli infection.
Trans R Soc Trop Med Hyg, 102 (2008), pp. 852-856
[113]
H.S. Maranhão, M.C.C. Medeiros, I.C.A. Scaletsky, U. Fagundes-Neto, M.B. Morais.
The epidemiological and clinical characteristics and nutritional development of infants with acute diarrhea, in northeastern Brazil.
Ann Trop Med Parasitol, 102 (2008), pp. 357-365
[114]
T.A.T. Gomes, M.A.M. Vieira, I.K. Wachsmuth, P.A. Blake, L.R. Trabulsi.
Serotype-specific prevalence of Escherichia coli strains with EPEC adherence factor genes in infants with and without diarrhea in São Paulo, Brazil.
J Infect Dis, 160 (1989), pp. 131-135
[115]
M.R. Franzolin, R.C.B. Alves, R. Keller, et al.
Prevalence of diarrheagenic Escherichia coli in children with diarrhea in Salvador, Bahia, Brazil.
Mem Inst Oswaldo Cruz, 100 (2005), pp. 359-363
[116]
M.Y. Alikhani, A. Mirsalehian, M.M. Aslani.
Detection of typical and atypical enteropathogenic Escherichia coli (EPEC) in Iranian children with and without diarrhea.
J Med Microbiol, 55 (2006), pp. 1159-1163
[117]
P. Rajendran, S.S.R. Ajjampur, D. Chidambaram, et al.
Pathotypes of diarrheagenic Escherichia coli in children attending a tertiary care hospital in South India.
Diagn Microbiol Infect Dis, 68 (2010), pp. 117-122
[118]
K.L. Kotloff, J.P. Nataro, W. Blackwelder, et al.
Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicentre Study, GEMS): a prospective, case–control study.
[119]
S. Santona, N. Diaz, P.L. Fiori, et al.
Genotypic and phenotypic features of enteropathogenic Escherichia coli isolated in industrialized and developing countries.
J Infect Dev Ctries, 7 (2013), pp. 214-219
[120]
B.S.B. Nejma, M. Hassine Zaafrane, F. Hassine, et al.
Etiology of acute diarrhea in tunisian children with emphasis on diarrheagenic Escherichia coli: prevalence and identification of E. coli virulence markers.
Iran J Public Health, 43 (2014), pp. 947-960
[121]
C. Langendorf, S. Le Hello, A. Moumouni, et al.
Enteric bacterial pathogens in children with diarrhea in Niger: diversity and antimicrobial resistance.
PLoS ONE, 10 (2015), pp. e0120275
[122]
B. Odetoyin, J. Hofmann, A. Aboderin, I. Okeke.
Diarrhoeagenic Escherichia coli in mother–child pairs in Ile-Ife, South Western Nigeria.
BMC Infect Dis, 28 (2016),
[123]
M.M. Levine, R. Edelman.
Enteropathogenic Escherichia coli of classic serotypes associated with infant diarrhea: epidemiology and pathogenesis.
Epidemiol Rev, 6 (1984), pp. 31-51
[124]
M.M. Levine, E.J. Bergquist, D.R. Nalin, et al.
Escherichia coli strains that cause diarrhea but do not produce heat-labile or heat-stable enterotoxins and are non-invasive.
Lancet, 1 (1978), pp. 1119-1122
[125]
I.K. Behiry, E.A. Abada, E.A. Ahmed, R.S. Labeed.
Enteropathogenic Escherichia coli associated with diarrhea in children in Cairo, Egypt.
Sci World J, 11 (2011), pp. 2613-2619
[126]
D.M. Lozer, T.B. Souza, M.V. Monfardini, et al.
Genotypic and phenotypic analysis of diarrheagenic Escherichia coli strains isolated from Brazilian children living in low socioeconomic level communities.
BMC Infect Dis, 13 (2013), pp. 418
[127]
F.E.A. Assis, S. Wolf, M. Surek, et al.
Impact of Aeromonas and diarrheagenic Escherichia coli screening in patients with diarrhea in Paraná, Southern Brazil.
J Infect Dev Ctries, 8 (2014), pp. 1609-1614
[128]
R.C.B. Dias, B.C. Santos, L.F. Santos, et al.
Diarrheagenic Escherichia coli pathotypes investigation revealed atypical enteropathogenic E. coli as putative emerging diarrheal agents in children living in Botucatu, São Paulo State, Brazil.
APMIS, 124 (2016), pp. 299-308
[129]
M.A. Foster, J. Iqbal, C. Zhang, et al.
Enteropathogenic and enteroaggregative E. coli in stools of children with acute gastroenteritis in Davidson County, Tennessee.
Diagn Microbiol Infect Dis, 83 (2015), pp. 319-324
[130]
R. Kolenda, M. Burdukiewicz, P. Schierack.
A systematic review and meta-analysis of the epidemiology of pathogenic Escherichia coli of calves and the role of calves as reservoirs for human pathogenic E. coli.
Front Cell Infect Microbiol, 5 (2015), pp. 23
[131]
R.A. Moura, M.P. Sircili, L. Leomil, et al.
Clonal Relationship among Atypical enteropathogenic Escherichia coli strains isolated from different animal species and humans.
Appl Environ Microbiol, 75 (2009), pp. 7399-7408
[132]
V.P. Gannon, M. Rashed, R.K. King, E.J. Thomas.
Detection and characterization of the eae gene of Shiga-like toxin-producing Escherichia coli using polymerase chain reaction.
J Clin Microbiol, 31 (1993), pp. 1268-1274
[133]
H. Karch, H. Böhm, H. Schmidt, F. Gunzer, S. Aleksic, J. Heesemann.
Clonal structure and pathogenicity of Shiga-like toxin-producing, sorbitol-fermenting Escherichia coli O157:H-.
J Clin Microbiol, 31 (1993), pp. 1200-1205
[134]
T.S. Gunzburg, G.N. Tornieporth, W.L. Riley.
Identification of enteropathogenic Escherichia coli by PCR-based detection of the bundle-forming pilus gene.
J Clin Microbiol, 33 (1995), pp. 1375-1377
[135]
J. Franke, S. Franke, H. Schmidt, et al.
Nucleotide sequence analysis of enteropathogenic Escherichia coli (EPEC) adherence factor probe and development of PCR for rapid detection of EPEC harboring virulence plasmids.
J Clin Microbiol, 32 (1994), pp. 2460-2463
[136]
T.E. Blank, H. Zhong, A.L. Bell, T.S. Whittam, M.S. Donnenberg.
Molecular variation among type IV pilin (bfpA) genes from diverse enteropathogenic Escherichia coli strains.
Infect Immun, 68 (2000), pp. 7028-7038
[137]
World Health Organization.
Programme for control of diarrhoeal diseases (CDD/83.3 Rev.1).
Manual for Laboratory Investigation of Acute Enteric Infections, World Health Organization, (1987), pp. 27
[138]
L.R. Trabulsi, L.C. Campos, T.S. Whittam, T.A.T. Gomes, J. Rodrigues, A.G. Gonçalves.
Traditional and non-traditional enteropathogenic Escherichia coli serogroups.
pp. 1-6
[139]
R.M.F. Piazza, C.M. Abe, D.S.P.Q. Horton, et al.
Detection and subtyping methods of diarrheagenic Escherichia coli strains.
Pathogenic Escherichia coli in Latin America, pp. 95-115
[140]
J.M. Nara, A.M. Cianciarullo, H.F. Culler, et al.
Differentiation of typical and atypical enteropathogenic Escherichia coli using colony immunoblot for detection of bundle-forming pilus expression.
J Appl Microbiol, 109 (2010), pp. 35-43
[141]
J.A. Girón, F. Qadri, T. Azim, K.J. Jarvis, J.B. Kaper, M.J. Albert.
Monoclonal antibodies specific for the bundle-forming pilus of enteropathogenic Escherichia coli.
Infect Immun, 63 (1995), pp. 4949-4952
[142]
J. Gismero-Ordoñez, M. Dall’agnol, L.R. Trabulsi, J.A. Girón.
Expression of the bundle-forming pilus by enteropathogenic Escherichia coli strains of heterologous serotypes.
J Clin Microbiol, 40 (2002), pp. 2291-2296
[143]
J. Adu-Bobie, G. Frankel, C. Bain, et al.
Detection of intimins α, β, γ, and δ, four intimin derivatives expressed by attaching and effacing microbial pathogens.
J Clin Microbiol, 36 (1998), pp. 662-668
[144]
M. Batchelor, S. Knutton, A. Caprioli, et al.
Development of a universal intimin antiserum and PCR primers.
J Clin Microbiol, 37 (1999), pp. 3822-3827
[145]
P.C.M. Koga, C.A. Menezes, F.A. Lima, et al.
Polyclonal anti-intimin antibody: immunological characterization and its use in EPEC diagnosis.
Braz J Microbiol, 34 (2003), pp. 5-7
[146]
M.A. Menezes, L.B. Rocha, P.C.M. Koga, et al.
Identification of enteropathogenic and enterohaemorrhagic Escherichia coli strains by immunoserological detection of intimin.
J Appl Microbiol, 108 (2010), pp. 878-887
[147]
M.A. Menezes, K.A. Aires, C.Y. Ozaki, et al.
Cloning approach and functional analysis of anti-intimin single-chain variable fragment (scFv).
BMC Research Notes, 4 (2011), pp. 30
[148]
A. Caravelli, D.E. Luz, F.B. Andrade, C.T. Moraes, A.Q. Maranhão, R.M. Piazza.
Sensitive and specific detection of enteropathogenic and enterohemorrhagic Escherichia coli using recombinant anti-intimin antibody by immunofluorescence assay.
Diagn Microbiol Infect Dis, 77 (2013), pp. 301-303
[149]
Y. Lu, C. Toma, Y. Honma, M. Iwanaga.
Detection of EspB using reversed passive latec agglutination: application to determination of enteropathogenic Escherichia coli.
Diagn Microbiol Infect Dis, 43 (2002), pp. 7-12
[150]
N. Nakasone, C. Toma, Y. Lu, M. Iwanaga.
Development of a rapid immunochromatographic test to identify enteropathogenic and enterohemorrhagic Escherichia coli by detecting EspB.
Diagn Microbiol Infect Dis, 57 (2007), pp. 21-25
[151]
L.B. Rocha, A.R. Santos, D.D. Munhoz, et al.
Development of a rapid agglutination latex test for diagnosis of enteropathogenic and enterohemorrhagic Escherichia coli infection in developing world: defining the biomarker, antibody and method.
PLoS Negl Trop Dis, 8 (2014), pp. e3150
[152]
A.R. Melton-Celsa.
Shiga toxin (Stx) classification, structure, and function.
Microbiol Spectr, 2 (2014),
EHEC-0024-2013
[153]
B.E.C. Guth, C.F. Picheth, T.A.T. Gomes.
Escherichia coli situation in Brazil.
Pathogenic Escherichia coli in Latin America, pp. 162-178
[154]
S.E. Majowicz, E. Scallan, A. Jones-Bitton, et al.
Global incidence of human Shiga toxin-producing Escherichia coli infections and deaths: a systematic review and knowledge synthesis.
Foodborne Pathog Dis, 6 (2014), pp. 447-455
[155]
J.B. Kaper, A.D. O’Brien.
Overview and historical perspectives.
Microbiol Spectr, 2 (2014),
EHEC-0028-2014
[156]
L.H. Gould, R.K. Mody, K.L. Ong, et al.
Increased recognition of non-O157 Shiga toxin-producing Escherichia coli infections in the United States during 2000–2010: epidemiologic features and comparison with E. coli O157 infections.
Food Pathog Dis, 10 (2013), pp. 453-460
[157]
M. Muniesa, J.A. Hammerl, S. Stefan Hertwig, B. Appel, H. Brüssow.
Shiga toxin-producing Escherichia coli O104:H4: a new challenge for microbiology.
Appl Env Microbiol, 78 (2012), pp. 4065-4073
[158]
S. Bletz, M. Bielaszewska, S.R. Leopold, et al.
Evolution of enterohemorrhagic Escherichia coli O26 based on single-nucleotide polymorphisms.
Genome Biol Evol, 5 (2013), pp. 1807-1816
[159]
M. Rivas, I. Chinen, B.E.C. Guth.
Enterohemorrhagic (Shiga toxin-producing) Escherichia coli.
Escherichia coli in the Americas, pp. 97-123
[160]
F. Scheutz, L.D. Teel, L. Beutin, et al.
Multicenter evaluation of a sequence-based protocol for subtyping Shiga toxins and standardizing Stx nomenclature.
J Clin Microbiol, 50 (2012), pp. 2951-2963
[161]
F. Scheutz.
Taxonomy meets public health: the case of Shiga toxin-producing Escherichia coli.
Microbiol Spectr, 2 (2014),
EHEC-0019-2013
[162]
A.K. Persad, J.T. LeJeune.
Animal reservoirs of Shiga toxin-producing Escherichia coli.
Microbiol Spectr, 2 (2014),
EHEC-0027-2014
[163]
A. Krüger, P.M.A. Lucchesi.
Shiga toxins and stx phages: highly diverse entities.
Microbiology, 161 (2015), pp. 451-462
[164]
M.P. Stevens, G.M. Frankel.
The locus of enterocyte effacement and associated virulence factors of enterohemorrhagic Escherichia coli.
Microbiol Spectr, 2 (2014),
EHEC-0007-2013
[165]
A.W. Paton, M.C. Woodrow, R. Doyle, et al.
Molecular characterization of a Shiga-toxigenic Escherichia coli O113:H21 strain lacking eae responsible for a cluster of cases of hemolytic-uremic syndrome.
J Clin Microbiol, 37 (1999), pp. 3357-3361
[166]
M. Bielaszewska, A. Mellmann, W. Zhang, et al.
Characterization of the E. coli strain associated with an outbreak of haemolytic uraemic syndrome in Germany, 2011: a microbiological study.
Lancet Infect Dis, 11 (2011), pp. 671-676
[167]
A. Pacheco, V. Sperandio.
Enteric pathogens exploit the microbiota-generated nutritional environment of the gut.
[168]
C.G. Moreira, V. Sperandio.
The Epinephrine/norepinephrine/autoinducer-3 interkingdom signaling system in Escherichia coli O157:H7.
Adv Exp Med Biol, 874 (2016), pp. 247-261
[169]
F.T. Biscola, C.M. Abe, B.E.C. Guth.
Determination of adhesin gene sequences in, and biofilm formation by, O157 and non-O157 Shiga toxin-producing Escherichia coli strains isolated from different sources.
Appl Environ Microbiol, 77 (2011), pp. 2201-2208
[170]
C. Matheus-Guimarães, E. Gonçalves, B.E.C. Guth.
Interactions of O157 and non-O157 Shiga toxin-producing Escherichia coli (STEC) recovered from bovine hide and carcass with human cells and abiotic surfaces.
Foodborne Pathog Dis, 3 (2014), pp. 248-255
[171]
F. Cordeiro, R.I.K. Silva, T.L.Z. Vargas-Stampe, A.M.F. Cerqueira, J.R.C. Andrade.
Cell invasion and survival of Shiga toxin-producing Escherichia coli within cultured human intestinal epithelial cells.
Microbiol, 159 (2013), pp. 1683-1694
[172]
L.F. Dos Santos.
Studies on the Virulence Potential and Phylogeny of O113:H21 Escherichia coli Strains.
Universidade Federal de São Paulo, (2011),
[PhD thesis]
[173]
A.G. Gonzalez, A.M. Cerqueira, B.E.C. Guth, et al.
Serotypes, virulence markers and cell invasion ability of Shiga toxin-producing Escherichia coli (STEC) strains isolated from healthy dairy cattle.
J Appl Microbiol, 121 (2016), pp. 1130-1143
[174]
P.C.H. Feng, S. Delannoy, D.W. Lacher, et al.
Genetic diversity and virulence potential of Shiga toxin-producing Escherichia coli O113:H21 strains isolated from clinical, environmental, and food sources.
Appl Environ Microbiol, 80 (2014), pp. 4757-4763
[175]
R.L. De Souza, J.T.A. Carvalhaes, L.S. Nishimura, M.C. Andrade, B.E.C. Guth.
Hemolytic uremic syndrome in pediatric intensive care units in São Paulo, Brazil.
Open Microbiol J, 5 (2011), pp. 76-82
[176]
M. Guirro, R.M.F. Piazza, R.L. de Souza, B.E.C. Guth.
Humoral immune response to Shiga Toxin 2 (Stx2) among Brazilian urban children with hemolytic uremic syndrome and healthy controls.
BMC Infect Dis, 14 (2014), pp. 320-325
[177]
L.F. Dos Santos, B.E.C. Guth, R.T. Hernandes, et al.
Shiga toxin-producing Escherichia coli in Brazil: human infections from 2007 to 2014.
9th Triennial International Symposium on Shiga Toxin (Verocytotoxin)-producing Escherichia coli (VTEC), Boston, vol. 87,
[178]
K.M.S. Lascowski, E.M. Gonçalves, P.P. Alvares, et al.
Prevalence and virulence profiles of Shiga toxin-producing Escherichia coli isolated from beef cattle in a Brazilian slaughterhouse.
Zoon Publ Health, 59 (2012), pp. 19-90
[179]
L.G. Beraldo, C.A. Borges, R.P. Maluta, M.V. Cardozo, E.C. Rigobelo, F.A. A’vila.
Detection of Shiga toxigenic (STEC) and enteropathogenic (EPEC) Escherichia coli in dairy buffalo.
Vet Microbiol, 170 (2014), pp. 162-166
[180]
F.H. Martins, B.E.C. Guth, R.M. Piazza, et al.
Diversity of Shiga toxin-producing Escherichia coli in sheep flocks of Paraná State, Southern Brazil.
Vet Microbiol, 175 (2015), pp. 150-156
[181]
R.P. Maluta, J.M. Fairbrother, A.E. Stella, E.C. Rigobelo, R. Martinez, F.A. A’vila.
Potentially pathogenic Escherichia coli in healthy, pasture-raised sheep on farms and at the abattoir in Brazil.
Vet Microbiol, 169 (2014), pp. 89-95
[182]
C.A. Borges, L.G. Beraldo, R.P. Maluta, et al.
Shiga toxigenic and atypical enteropathogenic Escherichia coli in the feces and carcasses of slaughtered pigs.
Foodborne Pathog Dis, 10 (2012), pp. 1-7
[183]
R.P. Martins, M.C. Silva, V. Dutra, L. Nakazato, D.S. Leite.
Preliminary virulence genotyping and phylogeny of Escherichia coli from the gut of pigs at slaughtering stage in Brazil.
Meat Sci, 93 (2013), pp. 437-440
[184]
R.M. Gioia-Di Chiacchio, M.P.V. Cunha, R.M. Sturn, et al.
Shiga toxin-producing Escherichia coli (STEC): zoonotic risks associated with psittacine pet birds in home environments.
Vet Microbiol, 184 (2016), pp. 27-30
[185]
L.F. Ribeiro, M.M.C. Barbosa, F.R. Pinto.
Shiga toxigenic and enteropathogenic Escherichia coli in water and fish from pay-to-fish ponds.
Lett Appl Microbiol, 62 (2015), pp. 216-220
[186]
F.H. Martins, B.E.C. Guth, R.M.F. Piazza, J. Blanco, J.S. Pelayo.
First description of a Shiga toxin-producing Escherichia coli O103:H2 strain isolated from sheep in Brazil.
J Infect Dev Ctries, 8 (2014), pp. 126-128
[187]
K.M.S. Lascowski, B.E.C. Guth, F.H. Martins, S.P.D. Rocha, K. Irino, J.S. Pelayo.
Shiga toxin-producing Escherichia coli in drinking water supplies of North Paraná State, Brazil.
J Appl Microbiol, 114 (2013), pp. 1230-1239
[188]
J. Puño-Sarmiento, L.E. Gazal, L.P. Medeiros, E.K. Nishio, R.K.T. Kobayashi, G. Nakazato.
Identification of diarrheagenic Escherichia coli strains from avian organic fertilizers.
Int J Environ Res Public Health, 11 (2014), pp. 8924-8939
[189]
A. Lucatelli.
Shiga Toxin-producing Escherichia coli in Ground Beef at Retail Level at São Paulo City, Brazil.
Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, (2012),
Ms Thesis
[190]
J.T.M. Peresi, I.A.Z.C. Almeida, T.M.I. Vaz, et al.
Search for diarrheagenic Escherichia coli in raw kibbe samples reveals the presence of Shiga toxin-producing strains.
Food Control, 63 (2016), pp. 165-170
[191]
S.A. Hoffmann, G.G. Pieretti, A. Fiorini, E.V. Patussi, R.F. Cardoso, J.M.G. Mikcha.
Shiga-toxin genes and genetic diversity of Escherichia coli isolated from pasteurized cow milk in Brazil.
J Food Sci, 79 (2014), pp. 1175-1180
[192]
B.R.C. Leite Junior, P.M. Oliveira, F.J.M. Silva.
Occurrence of Shiga toxin-producing Escherichia coli (STEC) in bovine feces, feed, water, raw milk, pasteurized milk, Minas Frescal cheese and ground beef samples collected in Minas Gerais, Brazil.
Int Food Res J, 21 (2014), pp. 2481-2486
[193]
P.A. Chapman, C.A. Siddons.
A comparison of immunomagnetic separation and direct culture for the isolation of verocytotoxin-producing Escherichia coli 0157 from cases of bloody diarrhoea, non-bloody diarrhoea and asymptomatic contacts.
J Med Microbiol, 44 (1996), pp. 267-271
[194]
C.A. Bopp, F.W. Brenner, P.I. Fields, J.G. Wells, N.A. Strockbine.
Escherichia, Shigella, and Salmonella.
Manual of Clinical Microbiology, 8th edition,
[195]
J. Konowalchuk, J.I. Speirs, S. Stavric.
Vero response to a cytotoxin of Escherichia coli.
Infect Immun, 18 (1977), pp. 775-779
[196]
M.A. Karmali, B.T. Steele, M. Petric, C. Lim.
Sporadic cases of hemolytic uremic syndrome associated with fecal cytotoxin and cytotoxin-producing Escherichia coli.
Lancet, 1 (1983), pp. 619-620
[197]
G.A. Leotta, I. Chinen, S. Epszteyn, et al.
Validation of a multiplex PCR for detection of Shiga toxin-producing Escherichia coli.
Rev Argent Microbiol, 37 (2005), pp. 1-10
[198]
Center of Disease Control of United States, Centers for Disease Control and Prevention.
Recommendations for diagnosis of Shiga toxin-producing Escherichia coli infections by clinical laboratories.
MMWR, 58 (2009), pp. 1-12
[199]
A. Donohue-Rolfe, M.A. Kelley, M. Bennish, G.T. Keush.
Enzyme-linked immunosorbent assay for Shigella toxin.
J Clin Microbiol, 24 (1986), pp. 65-68
[200]
U. Kongmuang, T. Honda, T. Miwatani.
Enzyme-linked immunosorbent assay to detect Shiga toxin of Shigella dysenteriae and related toxins.
J Clin Microbiol, 25 (1987), pp. 115-118
[201]
A.M.R. Mackenzie, P. Lebel, E. Orrbine, et al.
Sensitivities and specificities of premier E. coli O157 and premier EHEC enzyme immunoassays for diagnosis of infection with verotoxin (Shiga-like toxin) producing Escherichia coli.
J Clin Microbiol, 36 (1998), pp. 160811
[202]
T.J. Novick, J.A. Daly, S.L. Mottice, K.C. Carroll.
Comparison of sorbitol MacConkey agar and a two-step method which utilizes enzyme-linked immunosorbent assay toxin testing and a chromogenic agar to detect and isolate enterohemorrhagic Escherichia coli.
J Clin Microbiol, 38 (2000), pp. 547-551
[203]
L. Beutin, S. Zimmermann, K. Gleier.
Rapid detection and isolation of Shiga-like toxin (verocytotoxin)-producing Escherichia coli by direct testing of individual enterohemolytic colonies from washed sheep blood agar plates in the VTEC-RPLA assay.
Clin Microbiol, 34 (1996), pp. 2812-2814
[204]
L. Beutin, S. Zimmermann, K. Gleier.
Evaluation of the VTEC-Screen “Seiken” test for detection of different types of Shiga toxin (verotoxin)-producing Escherichia coli (STEC) in human stool samples.
Diagn Microbiol Infect Dis, 42 (2002), pp. 1-8
[205]
L. Beutin, H. Steinrück, G. Krause, et al.
Comparative evaluation of the Ridascreen® Verotoxin enzyme immunoassay for detection of Shiga-toxin producing strains of Escherichia coli (STEC) from food and other sources.
J Appl Microbiol, 102 (2007), pp. 630-639
[206]
L.H. Gould, C. Bopp, N. Strockbine, et al.
Recommendations for diagnosis of Shiga toxin-producing Escherichia coli infections by clinical laboratories.
MMWR Recomm Rep, 58 (2009), pp. 1-14
[207]
L.B. Rocha, D. Luz, C.T.P. Moraes, et al.
Interaction between Shiga toxin and monoclonal antibodies: binding characteristics and in vitro neutralizing abilities.
Toxins, 4 (2012), pp. 729-747
[208]
L.B. Rocha, R.M.F. Piazza.
Production of Shiga toxin by Shiga toxin-expressing Escherichia coli (STEC) in broth media: from divergence to definition.
Lett Appl Microbiol, 45 (2007), pp. 411-417
[209]
M.R.B. Mendes-Ledesma, L.B. Rocha, V. Bueris, et al.
Production and characterization of rabbit polyclonal sera against Shiga toxins stx1 and stx2 for detection of Shiga toxin producing.
Microbiol Immunol, 52 (2008), pp. 484-491
[210]
D. Luz, H. Chen, A.Q. Maranhão, L.B. Rocha, S. Sidhu, R.M.F. Piazza.
Development and characterization of recombinant antibody fragments that recognize and neutralize in vitro stx2 toxin from Shiga toxin-producing Escherichia coli.
PLoS ONE, 10 (2015), pp. 0120481
[211]
F. Navarro-Garcia, W.P. Elias.
Autotransporters and virulence of enteroaggregative E. coli.
Gut Microbes, 2 (2011), pp. 13-24
[212]
J.P. Nataro, J.B. Kaper, R. Robins-Browne, V. Prado, P. Vial, M.M. Levine.
Patterns of adherence of diarrheagenic Escherichia coli to HEp-2 cells.
Pediatr Infect Dis J, 6 (1987), pp. 829-831
[213]
D.A. Rasko, D.R. Webster, J.W. Sahl, et al.
Origins of the E. coli strain causing an outbreak of hemolytic-uremic syndrome in Germany.
N Engl J Med, 365 (2011), pp. 709-717
[214]
B. Hebbelstrup Jensen, K.E. Olsen, C. Struve, K.A. Krogfelt, A.M. Petersen.
Epidemiology and clinical manifestations of enteroaggregative Escherichia coli.
Clin Microbiol Rev, 27 (2014), pp. 614-630
[215]
A.A. Lima, R.L. Guerrant.
Persistent diarrhea in children: epidemiology, risk factors, pathophysiology, nutritional impact, and management.
Epidemiol Rev, 14 (1992), pp. 222-242
[216]
Z.D. Jiang, P.C. Okhuysen, D.C. Guo, et al.
Genetic susceptibility to enteroaggregative Escherichia coli diarrhea: polymorphism in the interleukin-8 promotor region.
J Infect Dis, 188 (2003), pp. 506-511
[217]
J.A. Mohamed, H.L. DuPont, Z.D. Jiang, et al.
A novel single-nucleotide polymorphism in the lactoferrin gene is associated with susceptibility to diarrhea in North American travelers to Mexico.
Clin Infect Dis, 44 (2007), pp. 945-952
[218]
J.A. Mohamed, H.L. DuPont, Z.D. Jiang, et al.
A single-nucleotide polymorphism in the gene encoding osteoprotegerin, an anti-inflammatory protein produced in response to infection with diarrheagenic Escherichia coli, is associated with an increased risk of nonsecretory bacterial diarrhea in North American travelers to Mexico.
J Infect Dis, 199 (2009), pp. 477-485
[219]
J.A. Mohamed, H.L. DuPont, J. Flores, et al.
Single nucleotide polymorphisms in the promoter of the gene encoding the lipopolysaccharide receptor CD14 are associated with bacterial diarrhea in US and Canadian travelers to Mexico.
Clin Infect Dis, 52 (2011), pp. 1332-1341
[220]
J.R. Czeczulin, T.S. Whittam, I.R. Henderson, F. Navarro-Garcia, J.P. Nataro.
Phylogenetic analysis of enteroaggregative and diffusely adherent Escherichia coli.
Infect Immun, 67 (1999), pp. 2692-2699
[221]
S. Suzart, B.E. Guth, M.Z. Pedroso, U.M. Okafor, T.A. Gomes.
Diversity of surface structures and virulence genetic markers among enteroaggregative Escherichia coli (EAEC) strains with and without the EAEC DNA probe sequence.
FEMS Microbiol Lett, 201 (2001), pp. 163-168
[222]
W.P. Elias, A.P. Uber, S.K. Tomita, L.R. Trabulsi, T.A. Gomes.
Combinations of putative virulence markers in typical and variant enteroaggregative Escherichia coli strains from children with and without diarrhoea.
Epidemiol Infect, 129 (2002), pp. 49-55
[223]
N. Boisen, F. Scheutz, D.A. Rasko, et al.
Genomic characterization of enteroaggregative Escherichia coli from children in Mali.
J Infect Dis, 205 (2012), pp. 431-444
[224]
M.A. Chattaway, C. Jenkins, D. Rajendram, et al.
Enteroaggregative Escherichia coli have evolved independently as distinct complexes within the E. coli population with varying ability to cause disease.
[225]
I.N. Okeke, F. Wallace-Gadsden, H.R. Simons, et al.
Multi-locus sequence typing of enteroaggregative Escherichia coli isolates from Nigerian children uncovers multiple lineages.
[226]
I.F. Lima, N. Boisen, S. Quetz Jda, et al.
Prevalence of enteroaggregative Escherichia coli and its virulence-related genes in a case-control study among children from north-eastern Brazil.
J Med Microbiol, 62 (2013), pp. 683-693
[227]
J.P. Nataro, Y. Deng, S. Cookson, et al.
Heterogeneity of enteroaggregative Escherichia coli virulence demonstrated in volunteers.
J Infect Dis, 171 (1995), pp. 465-468
[228]
B. Baudry, S.J. Savarino, P. Vial, J.B. Kaper, M.M. Levine.
A sensitive and specific DNA probe to identify enteroaggregative Escherichia coli, a recently discovered diarrheal pathogen.
J Infect Dis, 161 (1990), pp. 1249-1251
[229]
J. Sarantuya, J. Nishi, N. Wakimoto, et al.
Typical enteroaggregative Escherichia coli is the most prevalent pathotype among E. coli strains causing diarrhea in Mongolian children.
J Clin Microbiol, 42 (2004), pp. 133-139
[230]
N. Morin, A.E. Santiago, R.K. Ernst, S.J. Guillot, J.P. Nataro.
Characterization of the AggR regulon in enteroaggregative Escherichia coli.
Infect Immun, 81 (2013), pp. 122-132
[231]
M. Cobeljic, B. Miljkovic-Selimovic, D. Paunovic-Todosijevic, et al.
Enteroaggregative Escherichia coli associated with an outbreak of diarrhoea in a neonatal nursery ward.
Epidemiol Infect, 117 (1996), pp. 11-16
[232]
Y. Itoh, I. Nagano, M. Kunishima, T. Ezaki.
Laboratory investigation of enteroaggregative Escherichia coli O untypeable:H10 associated with a massive outbreak of gastrointestinal illness.
J Clin Microbiol, 35 (1997), pp. 2546-2550
[233]
D.B. Huang, Z.D. Jiang, H.L. Dupont.
Association of virulence factor-positive and -negative enteroaggregative Escherichia coli and occurrence of clinical illness in travelers from the United States to Mexico.
Am J Trop Med Hyg, 69 (2003), pp. 506-508
[234]
D.B. Huang, J.A. Mohamed, J.P. Nataro, H.L. DuPont, Z.D. Jiang, P.C. Okhuysen.
Virulence characteristics and the molecular epidemiology of enteroaggregative Escherichia coli isolates from travellers to developing countries.
J Med Microbiol, 56 (2007), pp. 1386-1392
[235]
J.P. Nataro, Y. Deng, D.R. Maneval, A.L. German, W.C. Martin, M.M. Levine.
Aggregative adherence fimbriae I of enteroaggregative Escherichia coli mediate adherence to HEp-2 cells and hemagglutination of human erythrocytes.
Infect Immun, 60 (1992), pp. 2297-2304
[236]
W.P. Elias Jr., J.R. Czeczulin, I.R. Henderson, L.R. Trabulsi, J.P. Nataro.
Organization of biogenesis genes for aggregative adherence fimbria II defines a virulence gene cluster in enteroaggregative Escherichia coli.
J Bacteriol, 181 (1999), pp. 1779-1785
[237]
C. Bernier, P. Gounon, C. Le Bouguenec.
Identification of an aggregative adhesion fimbria (AAF) type III-encoding operon in enteroaggregative Escherichia coli as a sensitive probe for detecting the AAF-encoding operon family.
Infect Immun, 70 (2002), pp. 4302-4311
[238]
N. Boisen, C. Struve, F. Scheutz, K.A. Krogfelt, J.P. Nataro.
New adhesin of enteroaggregative Escherichia coli related to the Afa/Dr/AAF family.
Infect Immun, 76 (2008), pp. 3281-3292
[239]
R. Jonsson, C. Struve, N. Boisen, et al.
Novel aggregative adherence fimbria variant of enteroaggregative Escherichia coli.
Infect Immun, 83 (2015), pp. 1396-1405
[240]
S. Suzart, T.A.T. Gomes, B.E. Guth.
Characterization of serotypes and outer membrane protein profiles in enteroaggregative Escherichia coli strains.
Microbiol Immunol, 43 (1999), pp. 201-205
[241]
C. Debroy, J. Yealy, R.A. Wilson, M.K. Bhan, R. Kumar.
Antibodies raised against the outer membrane protein interrupt adherence of enteroaggregative Escherichia coli.
Infect Immun, 63 (1995), pp. 2873-2879
[242]
V. Monteiro-Neto, S.Y. Bando, C.A. Moreira-Filho, J.A. Giron.
Characterization of an outer membrane protein associated with haemagglutination and adhesive properties of enteroaggregative Escherichia coli O111:H12.
Cell Microbiol, 5 (2003), pp. 533-547
[243]
A. Zamboni, S.H. Fabbricotti, U. Fagundes-Neto, I.C. Scaletsky.
Enteroaggregative Escherichia coli virulence factors are found to be associated with infantile diarrhea in Brazil.
J Clin Microbiol, 42 (2004), pp. 1058-1063
[244]
A.L. Pereira, L.R. Ferraz, R.S. Silva, L.G. Giugliano.
Enteroaggregative Escherichia coli virulence markers: positive association with distinct clinical characteristics and segregation into 3 enteropathogenic E. coli serogroups.
J Infect Dis, 195 (2007), pp. 366-374
[245]
A.H. Regua-Mangia, T.A. Gomes, M.A. Vieira, K. Irino, L.M. Teixeira.
Molecular typing and virulence of enteroaggregative Escherichia coli strains isolated from children with and without diarrhoea in Rio de Janeiro city, Brazil.
J Med Microbiol, 58 (2009), pp. 414-422
[246]
J. Sheikh, J.R. Czeczulin, S. Harrington, et al.
A novel dispersin protein in enteroaggregative Escherichia coli.
J Clin Invest, 110 (2002), pp. 1329-1337
[247]
J. Nishi, J. Sheikh, K. Mizuguchi, et al.
The export of coat protein from enteroaggregative Escherichia coli by a specific ATP-binding cassette transporter system.
J Biol Chem, 278 (2003), pp. 45680-45689
[248]
B.T. Monteiro, L.C. Campos, M.P. Sircili, et al.
The dispersin-encoding gene (aap) is not restricted to enteroaggregative Escherichia coli.
Diagn Microbiol Infect Dis, 65 (2009), pp. 81-84
[249]
S.J. Savarino, A. Fasano, D.C. Robertson, M.M. Levine.
Enteroaggregative Escherichia coli elaborate a heat-stable enterotoxin demonstrable in an in vitro rabbit intestinal model.
J Clin Invest, 87 (1991), pp. 1450-1455
[250]
S.J. Savarino, A. Fasano, J. Watson, et al.
Enteroaggregative Escherichia coli heat-stable enterotoxin 1 represents another subfamily of E. coli heat-stable toxin.
Proc Natl Acad Sci U S A, 90 (1993), pp. 3093-3097
[251]
A. Fasano, F.R. Noriega, F.M. Liao, W. Wang, M.M. Levine.
Effect of Shigella enterotoxin 1 (ShET1) on rabbit intestine in vitro and in vivo.
Gut, 40 (1997), pp. 505-511
[252]
L.P. Menard, J.G. Lussier, F. Lepine, C. Paiva de Sousa, J.D. Dubreuil.
Expression, purification, and biochemical characterization of enteroaggregative Escherichia coli heat-stable enterotoxin 1.
Protein Expr Purif, 33 (2004), pp. 223-231
[253]
C. Eslava, F. Navarro-Garcia, J.R. Czeczulin, I.R. Henderson, A. Cravioto, J.P. Nataro.
Pet, an autotransporter enterotoxin from enteroaggregative Escherichia coli.
Infect Immun, 66 (1998), pp. 3155-3163
[254]
I.R. Henderson, J. Czeczulin, C. Eslava, F. Noriega, J.P. Nataro.
Characterization of Pic, a secreted protease of Shigella flexneri and enteroaggregative Escherichia coli.
Infect Immun, 67 (1999), pp. 5587-5596
[255]
N. Dautin.
Serine protease autotransporters of enterobacteriaceae (SPATEs): biogenesis and function.
Toxins, 2 (2010), pp. 1179-1206
[256]
F. Navarro-Garcia, C. Eslava, J.M. Villaseca, et al.
In vitro effects of a high-molecular-weight heat-labile enterotoxin from enteroaggregative Escherichia coli.
Infect Immun, 66 (1998), pp. 3149-3154
[257]
F. Ruiz-Perez, J.P. Nataro.
Bacterial serine proteases secreted by the autotransporter pathway: classification, specificity, and role in virulence.
Cell Mol Life Sci, 71 (2014), pp. 745-770
[258]
A.G. Abreu, T.R. Fraga, A.P. Granados Martínez, et al.
The serine protease Pic from enteroaggregative Escherichia coli mediates immune evasion by the direct cleavage of complement proteins.
J Infect Dis, 212 (2015), pp. 106-115
[259]
E.M. Bellini, W.P. Elias, T.A. Gomes, et al.
Antibody response against plasmid-encoded toxin (Pet) and the protein involved in intestinal colonization (Pic) in children with diarrhea produced by enteroaggregative Escherichia coli.
FEMS Immunol Med Microbiol, 43 (2005), pp. 259-264
[260]
P.A. Vial, R. Robins-Browne, H. Lior, et al.
Characterization of enteroadherent-aggregative Escherichia coli, a putative agent of diarrheal disease.
J Infect Dis, 158 (1988), pp. 70-79
[261]
S.K. Tickoo, M.K. Bhan, R. Srivastava, et al.
Intestinal colonization and production of diarrhoea by enteroadherent-aggregative Escherichia coli.
Indian J Med Res, 95 (1992), pp. 278-283
[262]
S. Tzipori, J. Montanaro, R.M. Robins-Browne, P. Vial, R. Gibson, M.M. Levine.
Studies with enteroaggregative Escherichia coli in the gnotobiotic piglet gastroenteritis model.
Infect Immun, 60 (1992), pp. 5302-5306
[263]
J.J. Mathewson, P.C. Johnson, H.L. DuPont, T.K. Satterwhite, D.K. Winsor.
Pathogenicity of enteroadherent Escherichia coli in adult volunteers.
J Infect Dis, 154 (1986), pp. 524-527
[264]
R.R. Chaudhuri, M. Sebaihia, J.L. Hobman, et al.
Complete genome sequence and comparative metabolic profiling of the prototypical enteroaggregative Escherichia coli strain 042.
[265]
J.P. Nataro, M.M. Baldini, J.B. Kaper, R.E. Black, N. Bravo, M.M. Levine.
Detection of an adherence factor of enteropathogenic Escherichia coli with a DNA probe.
J Infect Dis, 152 (1985), pp. 560-565
[266]
S. Hicks, D.C. Candy, A.D. Phillips.
Adhesion of enteroaggregative Escherichia coli to pediatric intestinal mucosa in vitro.
Infect Immun, 64 (1996), pp. 4751-4760
[267]
J.P. Nataro, S. Hicks, A.D. Phillips, P.A. Vial, C.L. Sears.
T84 cells in culture as a model for enteroaggregative Escherichia coli pathogenesis.
Infect Immun, 64 (1996), pp. 4761-4768
[268]
C.M. Abe, S. Knutton, M.Z. Pedroso, E. Freymüller, T.A. Gomes.
An enteroaggregative Escherichia coli strain of serotype O111:H12 damages and invades cultured T84 cells and human colonic mucosa.
FEMS Microbiol Lett, 203 (2001), pp. 199-205
[269]
J.A. Andrade, E. Freymuller, U. Fagundes-Neto.
Pathophysiology of enteroaggregative Escherichia coli infection: an experimental model utilizing transmission electron microscopy.
Arq Gastroenterol, 47 (2010), pp. 306-312
[270]
J.A. Andrade, E. Freymuller, U. Fagundes-Neto.
Adherence of enteroaggregative Escherichia coli to the ileal and colonic mucosa: an in vitro study utilizing the scanning electron microscopy.
Arq Gastroenterol, 48 (2011), pp. 199-204
[271]
A.G. Torres, X. Zhou, J.B. Kaper.
Adherence of diarrheagenic Escherichia coli strains to epithelial cells.
Infect Immun, 73 (2005), pp. 18-29
[272]
F. Navarro-Garcia, J. Gutierrez-Jimenez, C. Garcia-Tovar, L.A. Castro, H. Salazar-Gonzalez, V. Cordova.
Pic, an autotransporter protein secreted by different pathogens in the Enterobacteriaceae family, is a potent mucus secretagogue.
Infect Immun, 78 (2010), pp. 4101-4109
[273]
S.M. Harrington, E.G. Dudley, J.P. Nataro.
Pathogenesis of enteroaggregative Escherichia coli infection.
FEMS MicrobioL Lett, 254 (2006), pp. 12-18
[274]
T.S. Steiner, A.A. Lima, J.P. Nataro, R.L. Guerrant.
Enteroaggregative Escherichia coli produce intestinal inflammation and growth impairment and cause interleukin-8 release from intestinal epithelial cells.
J Infect Dis, 177 (1998), pp. 88-96
[275]
Z.D. Jiang, D. Greenberg, J.P. Nataro, R. Steffen, H.L. DuPont.
Rate of occurrence and pathogenic effect of enteroaggregative Escherichia coli virulence factors in international travelers.
J Clin Microbiol, 40 (2002), pp. 4185-4190
[276]
D.B. Huang, H.L. DuPont, Z.D. Jiang, L. Carlin, P.C. Okhuysen.
Interleukin-8 response in an intestinal HCT-8 cell line infected with enteroaggregative and enterotoxigenic Escherichia coli.
Clin Diagn Lab Immunol, 11 (2004), pp. 548-551
[277]
Z. Benjelloun-Touimi, P.J. Sansonetti, C. Parsot.
SepA, the major extracellular protein of Shigella flexneri: autonomous secretion and involvement in tissue invasion.
Mol Microbiol, 17 (1995), pp. 123-135
[278]
K. Al-Hasani, I.R. Henderson, H. Sakellaris, et al.
The sigA gene which is borne on the she pathogenicity island of Shigella flexneri 2a encodes an exported cytopathic protease involved in intestinal fluid accumulation.
Infect Immun, 68 (2000), pp. 2457-2463
[279]
A. Mellmann, D. Harmsen, C.A. Cummings, et al.
Prospective genomic characterization of the German enterohemorrhagic Escherichia coli O104:H4 outbreak by rapid next generation sequencing technology.
[280]
D.B. Huang, J.P. Nataro, H.L. DuPont, et al.
Enteroaggregative Escherichia coli is a cause of acute diarrheal illness: a meta-analysis.
Clin Infect Dis, 43 (2006), pp. 556-563
[281]
N. Pabalan, E. Singian, H. Jarjanazi, T.S. Steiner.
Enteroaggregative Escherichia coli and acute diarrhea in children: a meta-analysis of South Asian populations.
Eur J Clin Microbiol Infect Dis, 32 (2013), pp. 597-607
[282]
J.P. Nataro, V. Mai, J. Johnson, et al.
Diarrheagenic Escherichia coli infection in Baltimore, Maryland, and New Haven, Connecticut.
Clin Infect Dis, 43 (2006), pp. 402-407
[283]
M.A. Chattaway, R. Harris, C. Jenkins, et al.
Investigating the link between the presence of enteroaggregative Escherichia coli and infectious intestinal disease in the United Kingdom, 1993 to 1996 and 2008 to 2009.
Euro Surveill, 18 (2013),
pii: 20582
[284]
C. Eslava, J. Villaseca, R. Morales, A. Navarro, A. Cravioto.
Identification of a protein with toxigenic activity produced by enteroaggregative Escherichia coli.
Abstracts of the 93rd General Meeting of the American Society for Microbiology,
[285]
M. Pai, G. Kang, B.S. Ramakrishna, A. Venkataraman, J. Muliyil.
An epidemic of diarrhoea in south India caused by enteroaggregative Escherichia coli.
Indian J Med Res, 106 (1997), pp. 7-12
[286]
G. Scavia, M. Staffolani, S. Fisichella, et al.
Enteroaggregative Escherichia coli associated with a foodborne outbreak of gastroenteritis.
J Med Microbiol, 57 (2008), pp. 1141-1146
[287]
A. Cravioto, A. Tello, A. Navarro, et al.
Association of Escherichia coli HEp-2 adherence patterns with type and duration of diarrhoea.
Lancet, 337 (1991), pp. 262-264
[288]
G.D. Fang, A.A. Lima, C.V. Martins, J.P. Nataro, R.L. Guerrant.
Etiology and epidemiology of persistent diarrhea in northeastern Brazil: a hospital-based, prospective, case–control study.
J Pediatr Gastroenterol Nutr, 21 (1995), pp. 137-144
[289]
C.A. Wanke, J.B. Schorling, L.J. Barrett, M.A. Desouza, R.L. Guerrant.
Potential role of adherence traits of Escherichia coli in persistent diarrhea in an urban Brazilian slum.
Pediatr Infect Dis J, 10 (1991), pp. 746-751
[290]
A.A. Lima, S.R. Moore, M.S. Barboza Jr., et al.
Persistent diarrhea signals a critical period of increased diarrhea burdens and nutritional shortfalls: a prospective cohort study among children in northeastern Brazil.
J Infect Dis, 181 (2000), pp. 1643-1651
[291]
J.A. Opintan, M.J. Newman, P.F. Ayeh-Kumi, et al.
Pediatric diarrhea in southern Ghana: etiology and association with intestinal inflammation and malnutrition.
Am J Trop Med Hyg, 83 (2010), pp. 936-943
[292]
J.K. Roche, A. Cabel, J. Sevilleja, J. Nataro, R.L. Guerrant.
Enteroaggregative Escherichia coli (EAEC) impairs growth while malnutrition worsens EAEC infection: a novel murine model of the infection malnutrition cycle.
J Infect Dis, 202 (2010), pp. 506-514
[293]
T.B. Morais, T.A.T. Gomes, D.M. Sigulem.
Enteroaggregative Escherichia coli in infant feeding bottles.
Lancet, 349 (1997), pp. 1448-1449
[294]
J.A. Adachi, J.J. Mathewson, Z.D. Jiang, C.D. Ericsson, H.L. DuPont.
Enteric pathogens in Mexican sauces of popular restaurants in Guadalajara, Mexico, and Houston, Texas.
Ann Intern Med, 136 (2002), pp. 884-887
[295]
A.P. Uber, L.R. Trabulsi, K. Irino, et al.
Enteroaggregative Escherichia coli from humans and animals differ in major phenotypical traits and virulence genes.
FEMS Microbiol Lett, 256 (2006), pp. 251-257
[296]
C.M. Abe, F.A. Salvador, I.N. Falsetti, et al.
Uropathogenic Escherichia coli (UPEC) strains may carry virulence properties of diarrhoeagenic E. coli.
FEMS Immunol Med Microbiol, 52 (2008), pp. 397-406
[297]
A.H. Regua-Mangia, K. Irino, R. da Silva Pacheco, R.M. Pimentel Bezerra, A.R. Santos Périssé, L.M. Teixeira.
Molecular characterization of uropathogenic and diarrheagenic Escherichia coli pathotypes.
J Basic Microbiol, 50 (2010), pp. S107-S115
[298]
H.K. Park, Y.J. Jung, H.C. Chae, et al.
Comparison of Escherichia coli uropathogenic genes (kps, usp and ireA) and enteroaggregative genes (aggR and aap) via multiplex polymerase chain reaction from suprapubic urine specimens of young children with fever.
Scand J Urol Nephrol, 43 (2009), pp. 51-57
[299]
A. Nazemi, M. Mirinargasi, N. Merikhi, S.H. Sharifi.
Distribution of pathogenic genes aatA, aap, aggR, among uropathogenic Escherichia coli (UPEC) and their linkage with stbA gene.
Indian J Microbiol, 51 (2011), pp. 355-358
[300]
K. Herzog, J. Engeler Dusel, M. Hugentobler, et al.
Diarrheagenic enteroaggregative Escherichia coli causing urinary tract infection and bacteremia leading to sepsis.
Infect, 42 (2014), pp. 441-444
[301]
T.A. Gomes, C.M. Abe, L.R. Marques.
Detection of HeLa cell-detaching activity and alpha-hemolysin production in enteroaggregative Escherichia coli strains isolated from feces of Brazilian children.
J Clin Microbiol, 33 (1995), pp. 3364
[302]
F. Wallace-Gadsden, J.R. Johnson, J. Wain, I.N. Okeke.
Enteroaggregative Escherichia coli related to uropathogenic clonal group A.
Emerg Infect Dis, 13 (2007), pp. 757-760
[303]
B. Olesen, F. Scheutz, R.L. Andersen, et al.
Enteroaggregative Escherichia coli O78:H10, the cause of an outbreak of urinary tract infection.
J Clin Microbiol, 50 (2012), pp. 3703-3711
[304]
E.J. Boll, C. Struve, N. Boisen, B. Olesen, S.G. Stahlhut, K.A. Krogfelt.
Role of enteroaggregative Escherichia coli virulence factors in uropathogenesis.
Infect Immun, 81 (2013), pp. 1164-1171
[305]
M.E. Rüttler, C.S. Yanzón, M.J. Cuitiño, N.F. Renna, M.A. Pizarro, A.M. Ortiz.
Evaluation of a multiplex PCR method to detect enteroaggregative Escherichia coli.
Biocell, 30 (2006), pp. 301-308
[306]
A.B. Vilhena-Costa, R.M.F. Piazza, J.M. Nara, L.R. Trabulsi, M.B. Martinez.
Slot blot immunoassay as a tool for plasmid-encoded toxin detection in enteroaggregative Escherichia coli culture supernatants.
Diagn Microbiol Infect Dis, 55 (2006), pp. 101-106
[307]
M.M. Levine, V. Prado, R. Robins-Browne, et al.
Use of DNA probes and HEp-2 cell adherence assay to detect diarrheagenic Escherichia coli.
J Infect Dis, 158 (1988), pp. 224-228
[308]
H. Schmidt, C. Knop, S. Franke, S. Aleksic, J. Heesemann, H. Karch.
Development of PCR for screening of enteroaggregative Escherichia coli.
J Clin Microbiol, 33 (1995), pp. 701-705
[309]
J.F. Cerna, J.P. Nataro, T. Estrada-Garcia.
Multiplex PCR for detection of three plasmid-borne genes of enteroaggregative Escherichia coli strains.
J Clin Microbiol, 41 (2003), pp. 2138-2140
[310]
F. Cordeiro, D.G. Pereira, M.R. Rocha, M.D. Asensi, W.P. Elias, L.C. Campos.
Evaluation of a multiplex PCR for identification of enteroaggregative Escherichia coli.
J Clin Microbiol, 46 (2008), pp. 828-829
[311]
J. Antikainen, E. Tarkka, K. Haukka, A. Siitonen, M. Vaara, J. Kirveskari.
New 16-plex PCR method for rapid detection of diarrheagenic Escherichia coli directly from stool samples.
Eur J Clin Microbiol, 28 (2009), pp. 899-908
[312]
S. Bouzari, A. Jafari, M. Zarepour.
Distribution of virulence related genes among enteroaggregative Escherichia coli isolates: using multiplex PCR and hybridization.
Infect Genet Evol, 5 (2005), pp. 79-83
[313]
C. Jenkins, H. Chart, G.A. Willshaw, T. Cheasty, H.R. Smith.
Genotyping of enteroaggregative Escherichia coli and identification of target genes for the detection of both typical and atypical strains.
Diagn Microbiol Infect Dis, 55 (2006), pp. 13-19
[314]
S. Panchalingam, M. Antonio, A. Hossain, et al.
Diagnostic microbiologic methods in the GEMS-1 case/control study.
Clin Infect Dis, 55 (2012), pp. S294-S302
[315]
E.G. Dudley, N.R. Thomson, J. Parkhill, N.P. Morin, J.P. Nataro.
Proteomic and microarray characterization of the AggR regulon identifies a pheU pathogenicity island in enteroaggregative Escherichia coli.
Mol Microbiol, 61 (2006), pp. 1267-1282
[316]
F.B. Andrade, T.A.T. Gomes, W.P. Elias.
A sensitive and specific molecular tool for detection of both typical and atypical enteroaggregative Escherichia coli.
J Microbiol Methods, 106 (2014), pp. 16-18
[317]
W. Gaastra, A.M. Svennerholm.
Colonization factors of human enterotoxigenic Escherichia coli (ETEC).
Trends Microbiol, 4 (1996), pp. 444-452
[318]
M.K. Wolf.
Occurrence, distribution, and association of O and H serogroups, colonization factor antigens, and toxins of enterotoxigenic Escherichia coli.
Clin Microbiol Rev, 10 (1997), pp. 569-584
[319]
B.E. Guth, A.B. Pacheco, W.M. von Krüger, L.C.S. Ferreira.
Comparison of outer membrane protein and lipopolysaccharide profiles of enterotoxigenic Escherichia coli strains isolated in São Paulo, Brazil.
Braz J Med Biol Res, 28 (1995), pp. 545-552
[320]
L.S. Nishimura, L.C.S. Ferreira, A.B.F. Pacheco, B.E. Guth.
Relationship between outer membrane protein and lipopolysaccharide profiles and serotypes of enterotoxigenic Escherichia coli isolated in Brazil.
FEMS Microbiol Lett, 143 (1996), pp. 253-258
[321]
F. Qadri, A.M. Svennerholm, A.S. Faruque, R.B. Sack.
Enterotoxigenic Escherichia coli in developing countries: epidemiology, microbiology, clinical features, treatment, and prevention.
Clin Microbiol Rev, 18 (2005), pp. 465-483
[322]
A.B.F. Pacheco, B.E.C. Guth, D.F. de Almeida, L.C.S. Ferreira.
Characterization of enterotoxigerfic Escherichia coli by random amplification of polymorphic DNA.
Res Microbiol, 147 (1996), pp. 175-182
[323]
A.B.F. Pacheco, B.E.C. Guth, K.C.C. Soares, D.F. de Almeida, L.C.S. Ferreira.
Clonal relationships among Escherichia coli serogroup O6 isolates based on RAPD.
FEMS Microbiol Lett, 148 (1997), pp. 255-260
[324]
A.B.F. Pacheco, B.E.C. Guth, K.C.C. Soares, L. Nishimura, D.F. de Almeida, L.C.S. Ferreira.
Random amplification of polymorphic DNA reveals serotype-specific clonal clusters among enterotoxigenic Escherichia coli strains isolated from humans.
J Clin Microbiol, 35 (1997), pp. 1521-1525
[325]
A.B.F. Pacheco, K.C.C. Soares, D.F. de Almeida, G.I. Viboud, N. Binsztein, L.C.S. Ferreira.
Clonal nature of enterotoxigenic Escherichia coli serotype O6:H16 revealed by randomly amplified polymorphic DNA analysis.
J Clin Microbiol, 36 (1998), pp. 2099-2102
[326]
A.B.F. Pacheco, L.C.S. Ferreira, M.G. Pichel, et al.
Beyond serotypes and virulence-associated factors: detection of genetic diversity among O153:H45 CFA/I heat-stable enterotoxigenic Escherichia coli strains.
J Clin Microbiol, 39 (2001), pp. 4500-4505
[327]
A.H. Regua-Mangia, B.C. Guth, J.R.C. Andrade, D.F. Almeida, N. Binsztein, G.I. Viboud.
Genotypic and phenotypic characterization of enterotoxigenic Escherichia coli (ETEC) strains isolated in Rio de Janeiro city, Brazil.
FEMS Immunol Med Microbiol, 40 (2004), pp. 155-162
[328]
H. Steinsland, D.W. Lacher, H. Sommerfelt, T.S. Whittam.
Ancestral lineages of human enterotoxigenic Escherichia coli.
J Clin Microbiol, 48 (2010), pp. 2916-2924
[329]
J.W. Sahl, D.A. Rasko.
Analysis of global transcriptional profiles of enterotoxigenic Escherichia coli isolate E24377A.
Infect Immun, 80 (2012), pp. 1232-1242
[330]
A. von Mentzer, T.R. Connor, L.H. Wieler, et al.
Identification of enterotoxigenic Escherichia coli (ETEC) clades with long-term global distribution.
Nat Genet, 46 (2014), pp. 1321-1326
[331]
S.D. Isidean, M.S. Riddle, S.J. Savarino, C.K. Porter.
A systematic review of ETEC epidemiology focusing on colonization factor and toxin expression.
Vaccine, 29 (2011), pp. 6167-6178
[332]
E. Joffré, A. Sjöling.
The LT1 and LT2 variants of the enterotoxigenic Escherichia coli (ETEC) heat-labile toxin (LT) are associated with major ETEC lineages.
Gut Microbes, 7 (2016), pp. 75-81
[333]
M.A. Lasaro, J.F. Rodrigues, C. Mathias-Santos, et al.
Genetic diversity of heat-labile toxin expressed by enterotoxigenic Escherichia coli strains isolated from humans.
J Bacteriol, 190 (2008), pp. 2400-2410
[334]
R.A. Nada, H.I. Shaheen, S.B. Khalil, et al.
Discovery and phylogenetic analysis of novel members of class b enterotoxigenic Escherichia coli adhesive fimbriae.
J Clin Microbiol, 49 (2011), pp. 1403-1410
[335]
A. von Mentzer.
Whole Genome Sequencing of Enterotoxigenic Escherichia coli (ETEC): Identification of ETEC Lineages and Novel Colonization Factors.
Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, (2016),
Thesis
[336]
T. Taniguchi, Y. Fujino, K. Yamamoto, T. Honda.
Sequencing of the gene encoding the major pilin of pilus colonization factor antigen III (CFA/III) of human enterotoxigenic Escherichia coli and evidence that CFA/III is related to type IV pili.
Infect Immun, 63 (1995), pp. 724-728
[337]
M. Pichel, N. Binsztein, G. Viboud.
CS22, a novel human enterotoxigenic Escherichia coli adhesin, is related to CS15.
Infect Immun, 68 (2000), pp. 3280-3285
[338]
F. Del Canto, D.J. Botkin, P. Valenzuela, et al.
Identification of coli surface antigen 23, a novel adhesin of enterotoxigenic Escherichia coli.
Infect Immun, 80 (2012), pp. 2791-2801
[339]
D.H. Francis.
Enterotoxigenic Escherichia coli infection in pigs and its diagnosis.
J Swine Health Prod, 10 (2002), pp. 171-175
[340]
J.M. Fleckenstein, P.R. Hardwidge, G.P. Munson, D.A. Rasko, H. Sommerfelt, H. Steinsland.
Molecular mechanisms of enterotoxigenic Escherichia coli infection.
Microbes Infect, 12 (2010), pp. 89-98
[341]
B.D. Spangler.
Structure and function of cholera toxin and the related Escherichia coli heat-labile enterotoxin.
Microbiol Rev, 56 (1992), pp. 622-647
[342]
T. Yamamoto, T. Yokota.
Sequence of heat-labile enterotoxin of Escherichia coli pathogenic for humans.
J Bacteriol, 155 (1983), pp. 728-733
[343]
P.A. Grange, L.A. Parrish, A.K. Erickson.
Expression of putative Escherichia coli heat-labile enterotoxin (LT) receptors on intestinal brush borders from pigs of different ages.
Vet Res Commun, 30 (2006), pp. 57-71
[344]
B.E. Guth, C.L. Pickett, E.M. Twiddy, et al.
Production of type II heat-labile enterotoxin by Escherichia coli isolated from food and human feces.
Infect Immun, 54 (1986), pp. 587-589
[345]
B.E. Guth, E.M. Twiddy, L.R. Trabulsi, R.K. Holmes.
Variation in chemical properties and antigenic determinants among type II heat-labile enterotoxins of Escherichia coli.
Infect Immun, 54 (1986), pp. 529-536
[346]
G. Hajishengallis, S. Arce, C.M. Gockel, T.D. Connell, M.W. Russell.
Immunomodulation with enterotoxins for the generation of secretory immunity or tolerance: applications for oral infections.
Crit Rev Oral Biol Med, 84 (2005), pp. 1104-1116
[347]
H.F. Nawar, N.D. King-Lyons, J.C. Hu, R.C. Pasek, T.D. Connell.
LT-IIc, a new member of the type II heat-labile enterotoxin family encoded by an Escherichia coli strain obtained from a non-mammalian host.
Infect Immun, 78 (2010), pp. 4705-4713
[348]
M.G. Jobling, R.K. Holmes.
Type II heat-labile enterotoxins from 50 diverse Escherichia coli isolates belong almost exclusively to the LT-IIc family and may be prophage encoded.
[349]
M.A. Lasaro, C. Mathias-Santos, J.F. Rodrigues, L.C.S. Ferreira.
Functional and immunological characterization of a natural polymorphic variant of a heat-labile toxin (LT-I) produced by enterotoxigenic Escherichia coli (ETEC).
FEMS Immunol Med Microbiol, 55 (2009), pp. 93-99
[350]
J.F. Rodrigues, C. Mathias-Santos, M.E. Sbrogio-Almeida, et al.
Functional diversity of heat-labile toxins (LT) produced by enterotoxigenic Escherichia coli: differential enzymatic and immunological activities of LT1 (hLT) and LT4 (pLT).
J Biol Chem, 286 (2011), pp. 5222-5233
[351]
E. Joffre, A. von Mentzer, M. Abd El Ghany, et al.
Allele variants of enterotoxigenic Escherichia coli heat-labile toxin are globally transmitted and associated with colonization factors.
J Bacteriol, 197 (2015), pp. 392-403
[352]
C. Zhang, D. Rausch, W. Zhang.
Little heterogeneity among genes encoding heat-labile and heat-stable toxins of enterotoxigenic Escherichia coli strains isolated from diarrheal pigs.
Appl Environ Microbiol, 75 (2009), pp. 6402-6405
[353]
E. Joffré, A. von Mentzer, A.M. Svennerholm, Å. Sjöling.
Identification of new heat-stable (STa) enterotoxin allele variants produced by human enterotoxigenic Escherichia coli (ETEC).
Int J Med Microbiol, 306 (2016), pp. 586-594
[354]
M.A.S. Lasaro, J.F. Rodrigues, C. Mathias-Santos, et al.
Production and release of heat-labile toxin by wild-type human-derived enterotoxigenic Escherichia coli.
FEMS Immunol Med Microbiol, 48 (2006), pp. 123-131
[355]
M.A.S. Lasaro, J.F. Rodrigues, J. Cabrera-Crespo, M.E. Sbrogio-Almeida, M.O. Lasaro, L.C.S. Ferreira.
Evaluation of experimental conditions for quantification of lt produced by human derived enterotoxigenic Escherichia coli strains.
Braz J Microbiol, 38 (2007), pp. 446-451
[356]
L.B. Rocha, C.Y. Ozaki, D.S. Horton, et al.
Different assay conditions for detecting the production and release of heat-labile and heat-stable toxins in enterotoxigenic Escherichia coli isolates.
Toxins, 5 (2013), pp. 2384-2402
[357]
L.M. Lamberti, A.L. Bourgeois, C.L.F. Walker, R.E. Black, D. Sack.
Estimating diarrheal illness and deaths attributable to Shigellae and enterotoxigenic Escherichia coli among older children, adolescents, and adults in South Asia and Africa.
PLoS Negl Trop Dis, 8 (2014), pp. e2705
[358]
M.H.L. Reis, A.F.P. Castro, M.R.F. Toledo, L.R. Trabulsi.
Production of heat-stable enterotoxin by the 0128 serogroup of Escherichia coli.
Infect Immun, 24 (1979), pp. 289-290
[359]
M.S.V. Gatti, L.C. Ricci, M.B. Serafim, A.F. De Castro.
Incidência de Escherichia coli enterotoxigênica (ETEC), rotavirus e Clostridium perfringens de casos de diarreia em crianças, na região de Campinas, SP, Brasil.
Rev Inst Med Trop S Paulo, 31 (1989), pp. 392-398
[360]
T.A.T. Gomes, V. Rassi, K.L. Mac Donald, et al.
Enteropathogens associated with acute diarrheal disease in urban infants in São Paulo, Brazil.
J Infect Dis, 164 (1991), pp. 331-337
[361]
A.H. Regua-Mangia, T.A.T. Gomes, M.A.M. Vieira, J.R. Andrade, K. Irino, L.M. Teixeira.
Frequency and characteristics of diarrhoeagenic Escherichia coli strains isolated from children with and without diarrhoea in Rio de Janeiro, Brazil.
J Infect, 48 (2004), pp. 161-167
[362]
M.H.L. Reis, B.E.C. Guth, T.A.T. Gomes, J. Murahovschi, L.R. Trabulsi.
Frequency of Escherichia coli strains producing heat-labile toxin or heat-stable toxin or both in children with and without diarrhea in Sao Paulo.
J Clin Microbiol, 15 (1982), pp. 1062-1064
[363]
M.E. Torres, M.C. Pírez, F. Schelotto, et al.
Etiology of children's diarrhea in Montevideo, Uruguay: associated pathogens and unusual isolates.
J Clin Microbiol, 39 (2001), pp. 2134-2139
[364]
R.B. Sack.
The discovery of cholera – like enterotoxins produced by Escherichia coli causing secretory diarrhoea in humans.
Indian. J. Med. Res, 133 (2011), pp. 171-178
[365]
S. Stacy-Phipps, J.J. Mecca, J.B. Weiss.
Multiplex PCR assay and simple preparation method for stool specimes detect enterotoxigenic Escherichia coli DNA during course of infection.
J Clin Microbiol, 33 (1995), pp. 1054-1059
[366]
B.P. Youmans, N.J. Ajami, Z.D. Jiang, J.F. Petrosino, H.L. DuPont, S.K. Highlander.
Development and accuracy of quantitative real-time polymerase chain reaction assays for detection and quantification of enterotoxigenic Escherichia coli (ETEC) heat labile and heat stable toxin genes in travelers’ diarrhea samples.
Am J Trop Med Hyg, 90 (2014), pp. 124-132
[367]
C. Toma, Y. Lu, N. Higa, et al.
Multiplex PCR assay for identification of human diarrheagenic Escherichia coli.
J Clin Microbiol, 41 (2003), pp. 2669-2671
[368]
R. Vidal, M. Vidal, R. Lagos, M. Levine, V. Prado.
Multiplex PCR for diagnosis of enteric infetions associated with diarrheagenic Escherichia coli.
J Clin Microbiol, 42 (2004), pp. 1787-1789
[369]
K.R. Aranda, S.H. Fabbricott, U. Fagundes-Neto, I.C. Scaletsky.
Single multiplex assay to identify simultaneously enteropathogenic, enteroaggregative, enterotoxigenic, enteroinvasive, and Shiga toxin-producing Escherichia coli strains in Brazilian children.
FEMS Microbiol Lett, 267 (2007), pp. 145-150
[370]
D.G. Evans, D.J. Evans Jr., N.F. Pierce.
Differences in the response of rabbit small intestine to heat-labile and heat-stable enterotoxins of Escherichia coli.
Infect Immun, 7 (1973), pp. 873-880
[371]
A.G. Dean, Y.C. Ching, R.G. Williams, L.B. Harden.
Test for Escherichia coli enterotoxin using infant mice: application in a study of diarrhea in children in Honolulu.
J Infect Dis, 125 (1972), pp. 407-411
[372]
S.T. Donta, H.W. Moon, S.C. Whipp.
Detection of heat-labile Escherichia coli enterotoxin with the use of adrenal cells in tissue culture.
Science, 183 (1974), pp. 334-336
[373]
R.L. Guerrant, L.L. Brunton, T.C. Schnaitman, L.I. Rebhun, A.G. Gilman.
Cyclic adenosine monophosphate and alteration of Chinese hamster ovary cell morphology: a rapid, sensitive in vitro assay for the enterotoxins of Vibrio cholerae and Escherichia coli.
Infect Immun, 10 (1974), pp. 320-327
[374]
R.A. Giannella, K.W. Drake, M. Luttrell.
Development of a radioimmunoassay for Escherichia coli heat-stable enterotoxin.
Infect Immun, 33 (1981), pp. 186-192
[375]
B. Cryan.
Comparison of three assay systems for detection of enterotoxigenic Escherichia coli heat-stable enterotoxin.
J Clin Microbiol, 28 (1990), pp. 792-794
[376]
E. Bläck, A.M. Svennerholm, J. Holmgren, R. Möllby.
Evaluation of a ganglioside immunosorbent assay for detection of Escherichia coli heat-labile enterotoxin.
J Clin Microbiol, 10 (1979), pp. 791-795
[377]
C.A. Menezes, S.Y. Imamura, L.R. Trabulsi, et al.
Production, characterization, and application of antibodies against heat-labile type-I toxin for detection of enterotoxigenic Escherichia coli.
Mem Inst Oswaldo Cruz, 101 (2006), pp. 875-880
[378]
C.A. Menezes, D.S. Gonçalves, J. Amianti, et al.
Capture immunoassay for LT detection produced by enterotoxigenic Escherichia coli in bacterial isolates.
Braz J Microbiol, 34 (2003), pp. 11-13
[379]
C.Y. Ozaki, C.R.F. Silveira, F.B. Andrade, et al.
Single chain variable fragments produced in Escherichia coli against heat-labile and heat-stable toxins from enterotoxigenic E. coli.
PLOS ONE, 10 (2015), pp. e0131484
[380]
L.R. Trabulsi, M.R.F. Toledo, J. Murahovschi, U. Fagundes Neto, J.A.M. Candeias.
Epidemiology of diarrhoea disease in South America.
Infectious Diarrhoea in the Young, pp. 121-125
[381]
R.M. Silva, M.R.F. Toledo, L.R. Trabulsi.
Biochemical and cultural characteristics of invasive Escherichia coli.
J Clin Microbiol, 11 (1980), pp. 441-444
[382]
S.B. Formal, R.B. Hornik.
Invasive Escherichia coli.
J Infect Dis, 137 (1978), pp. 641-644
[383]
S.B. Formal, T.L. Hale, P.J. Sansonetti.
Invasive enteric pathogens.
Rev Infect Dis, 5 (1983), pp. 702-707
[384]
W.H. Ewing, J.L. Gravatti.
Shigella types encountered in the mediterranean area.
J Bacteriol, 53 (1947), pp. 191-195
[385]
L.R. Trabulsi, M.R.F. Fernandes.
Antigenic identity of culture 193T-64 and E.coli O136:K78 (B22).
Rev Inst Med Trop, 11 (1969), pp. 101-103
[386]
R. Sakazaki, K. Tamura, M. Saito.
Enteropathogenic Escherichia coli associated with diarrhea in children and adults.
Jpn J Med Sci Biol, 20 (1967), pp. 387-399
[387]
I. Orskov, K. Wachsmuth, D.N. Taylor.
Two new Escherichia coli O groups: O172 from ‘shiga-like’ toxin II-producing strains (EHEC) and O173 from enteroinvasive E. coli (EIEC).
APMIS, 99 (1991), pp. 30-32
[388]
S. Matsushita, S. Yamada, A. Kai, Y. Kudoh.
Invasive strains of Escherichia coli belonging to serotype O121:NM.
J Clin Microbiol, 31 (1993), pp. 3034-3035
[389]
S. Vöros, B. Rédey, F. Csizmazia.
Antigenic structure of a new enteropathogenic Escherichia coli strains.
Acta Microbiol Acad Sci Hung, 11 (1964), pp. 125-129
[390]
M.R.F. Toledo, R.M. Silva, L.R. Trabulsi.
Sachs “Enterobacterium A12” is an aerogenic variant of Shigella boydii 14.
Int J Syst Bacteriol, 31 (1981), pp. 242-244
[391]
T.A.T. Gomes, M.R.F. Toledo, L.R. Trabulsi, P.K. Wood, J.G. Morris Jr..
DNA probes for identification of enteroinvasive Escherichia coli.
J Clin Microbiol, 25 (1987), pp. 2025-2027
[392]
V. Michelacci, G. Prosseda, A. Maugliani, et al.
Characterization of an emergent clone of enteroinvasive Escherichia coli circulating in Europe.
Clin Microbiol Infect, 22 (2016),
287.e11–9
[393]
S. Newitt, V. MacGregor, V. Robbins, et al.
Two linked enteroinvasive Escherichia coli outbreaks, Nottingham, UK, June 2014.
Emerg Infect Dis, 22 (2016), pp. 1178-1184
[394]
A. Andrade, J.A. Girón, J.M.K. Amhaz, L.R. Trabulsi, M.B. Martinez.
Expression and characterization of flagella in nonmotile enteroinvasive Escherichia coli isolated from diarrhea cases.
Infect Immun, 70 (2002), pp. 5882-5886
[395]
J.M.K. Amhaz, A. Andrade, S.Y. Bando, T.L. Tanaka, C.A. Moreira-Filho, M.B. Martinez.
Molecular typing and phylogenetic analysis of enteroinvasive Escherichia coli using the fliC gene sequence.
FEMS Microbiol Lett, 235 (2004), pp. 259-264
[396]
M.M. Levine.
E. coli that cause diarrhea enterotoxigenic, enteropathogenic, enteroinvasive, enterohemorragic and enteroaderent.
J Infect Dis, 155 (1987), pp. 377-389
[397]
P.J. Sansonetti, D.J. Kopecko, S.B. Formal.
Involvement of a palsmid in the invasive ability of Shigella flexneri.
Infect Immun, 35 (1982), pp. 852-860
[398]
S.B. Formal, T.L. Hale, P.J. Sansonetti.
Invasive enteric pathogens.
Rev Infect Dis, 5 (1983), pp. 702-707
[399]
J.R. Harris, I.K. Wachsmuth, B.R. Davis, M.L. Cohen.
High-molecular-weight plasmid correlates with Escherichia coli enteroinvasiveness.
Infect Immun, 37 (1982), pp. 1295-1298
[400]
A. Gibotti, T.L. Tanaka, V.R. Oliveira, C.R. Taddei, M.B. Martinez.
Molecular characterization of enteroinvasive Escherichia coli ipa genes by PCR-RFLP analysis.
Braz J Microbiol, 35 (2004), pp. 74-80
[401]
P.L.C. Small, R.R. Isberg, S. Falkow.
Comparation of the ability of enteroinvasive E.coli, Salmonella typhimuriun, Y. pseudotuberculosis, and Y. enterocolitica to enter and replicate within Hep-2 cells.
Infect Immun, 55 (1987), pp. 1674-1679
[402]
P. Cossart, P.J. Sansonetti.
Bacterial invasion: the paradigm of enteroinvasive pathogens.
Science, 304 (2004), pp. 242-248
[403]
C. Parsot.
Shigella spp. and enteroinvasive Escherichia coli pathogenicity factors.
FEMS Microbiol Lett, 252 (2005), pp. 11-18
[404]
M. Ogawa, Y. Handa, H. Ashida, M. Suzuki, C. Sasakawa.
The versatility of Shigella effectors.
Nat Rev Microbiol, 6 (2008), pp. 11-16
[405]
S.Y. Bando, G.R.F. Valle, M.B. Martinez, L.R. Trabulsi, C.A. Moureira-Filho.
Characterization of enteroinvasive Escherichia coli and Shigella strains by RAPD analysis.
FEMS Microbiol Lett, 165 (1998), pp. 159-165
[406]
R. Lan, M.C. Alles, K. Donohoe, M.B. Martinez, P.R. Reeves.
Molecular evolutionary relationships of enteroinvasive Escherichia coli and Shigella spp.
Infect Immun, 72 (2004), pp. 5080-5088
[407]
M.B. Martinez, T.S. Whittan, E.A. McGraw, J. Rodrigues, L.R. Trabulsi.
Clonal relationship among invasive and non-invasive strains of enteroinvasive Escherichia coli serogroups.
FEMS Microbiol Lett, 172 (1999), pp. 145-151
[408]
G.M. Pupo, D.K.R. Karaolis, R. Lan, P.R. Reeves.
Evolutionary relationships among pathogenic Escherichia coli strains inferred from multilocus enzyme electrophoresis and mdh sequence studies.
Infect Immun, 65 (1997), pp. 2685-2692
[409]
K. Rolland, N. Lambert-Zechovsky, B. Picard, E. Denamur.
Shigella and enteroinvasive Escherichia coli strains are derived from distinct ancestral strains of E. coli.
Microbiology, 144 (1998), pp. 2667-2672
[410]
H.L. Dupont, S.B. Formal, R.B. Hornick, et al.
Phatogenesis of Escherichia coli diarrhea.
N Engl J Med, 285 (1971), pp. 1-9
[411]
A.C. Moreno, L.G. Ferreira, M.B. Martinez.
Enteroinvasive Escherichia coli vs. Shigella flexneri: how different patterns of gene expression affect virulence.
FEMS Microbiol Lett, 301 (2009), pp. 156-163
[412]
A. Andrade, M. Dall’Agnol, S. Newton, M.B. Martinez.
The iron uptake mechanisms of enteroinvasive Escherichia coli.
Braz J Microbiol, 31 (2000), pp. 200-205
[413]
M. Dall’Agnol, M.B. Martinez.
Iron uptake from host compounds by enteroinvasive Escherichia coli.
Rev Microbiol, 30 (1999), pp. 149-152
[414]
P.J. Sansonetti, A.M. Phalipon.
Cells as ports of entry for enteroinvasive pathogens: mechanisms of interaction, consequences for the disease process.
Semin Immunol, 11 (1999), pp. 193-203
[415]
S.Y. Bando, A.C.R. Moreno, J.A.T. Albuquerque, J.M.K. Amhaz, C.A. Moureira-Filho, M.B. Martinez.
Expression of bacterial virulence factors and cytokines during in vitro macrophage infection by enteroinvasive Escherichia coli and Shigella flexneri: a comparative study.
Mem Inst Oswaldo Cruz, 105 (2010), pp. 1-6
[416]
A.C. Moreno, K.S. Ferreira, L.G. Ferreira, S.R. Almeida, M.B. Martinez.
Recognition of enteroinvasive Escherichia coli and Shigella flexneri by dendritic cells: distinct dendritic cell activation states.
Mem Inst Oswaldo Cruz, 107 (2012), pp. 138-141
[417]
L.R. Trabulsi, M.R.F. Fernandes, M.E. Zuliani.
Novas bactérias patogênicas para o intestino do homem.
Rev Inst Med Trop, 9 (1967), pp. 31-39
[418]
A. Borian, F. Csizmazia, E. Karvaly, F. Mihalffy, B. Redey.
Enterocolitis epidemic caused by water contamined with E. coli O124 in Veszpren.
Orv Hetil, 100 (1959), pp. 1072-1074
[419]
R. Marier, J.C. Wells, R.C. Swanson, W. Callahan, I.J. Mehlman.
An outbreak of enteropathogenic E. coli foodborne disease traced to imported cheese.
Lancet, 302 (1973), pp. 1376-1378
[420]
J.E.F. Tulloch, K.J. Ryan, S.B. Formal, F.A. Franklin.
Invasive enterophatic Escherichia coli dysentery.
Ann Intern Med, 79 (1973), pp. 13-17
[421]
S.R. Valentini, T.A.T. Gomes, D.P. Falcão.
Lack of virulence factors in Escherichia coli strains of enteropathogenic serougroups isolated from water.
Appl Environ Microbiol, 58 (1992), pp. 412-414
[422]
J.R. Harris, J. Mariano, J.G. Wells, B.J. Payne, H.D. Donnell, M.L. Cohen.
Person-to-person transmission in an outbreak of enteroinvasive Escherichia coli.
Am J Epidemiol, 122 (1985), pp. 245-252
[424]
B.D. Chaterjee, S.N. Sanyal.
Is it all shigellosis?.
[425]
S. Ram, S. Khurana, S.B. Khurana, S. Sharma, D.V. Vadehra.
Seasonal fluctuations in the occurrence of enteroinvasive Escherichia coli diarrhea.
Indian J Med Res Sec A, 91 (1990), pp. 258-262
[426]
P. Echeverria, O. Sethabutr, O. Serichantalergs, U. Lexomboon, K. Tamura.
Shigella and enteroinvasive Escherichia coli infections in households of children with dysentery in Bangkok.
J Infect Dis, 165 (1992), pp. 144-147
[427]
K.C. Kain, R.L. Barteluk, M.T. Kelly, et al.
Etiology of childhood diarrhea in Beijing, China.
J Clin Microbiol, 29 (1991), pp. 90-95
[428]
K. Tamura, R. Sakazaki, M. Murase, Y. Kosako.
Serotyping and categorisation of Escherichia coli strains isolated between 1958 and 1992 from diarrhoeal diseses in Asia.
J Med Microbiol, 45 (1996), pp. 353-358
[429]
D.N. Taylor, P. Escheverria, T. Pál, et al.
The role of Shigella sp, enteroinvasive Escherichia coli, and other enteropathogens as causes of childhood dysentery in Thailand.
J Infect Dis, 153 (1986), pp. 1132-1138
[430]
A. Utsunomiya, D. Elio-Calvo, A.A. Reyes, et al.
Major enteropathogenic bacteria isolated from diarrheal patients in Bolivia: a hospital-based study.
Microbiol Immunol, 39 (1995), pp. 845-851
[431]
M. Katouli, A. Jaari, A.A. Moghaddam, G.R. Ketabi.
Etiological studies of diarrheal diseases in infants and young children in Iran.
J Trop Med Hyg, 93 (1990), pp. 22-27
[432]
T.I. Ogunsanya, V.O. Rotimi, A. Adenuga.
A study of aetiological agents of childhood diarrhoea in Lagos, Nigeria.
J Med Microbiol, 40 (1994), pp. 10-14
[433]
G. Prats, T. Llovet.
Enteroinvasive Escherichia coli: pathogenic mechanisms and epidemiology.
Microbiologia, 11 (1995), pp. 91-96
[434]
R. Sunthadvanich, D. Chiewsilp, J. Seriwatana, R. Sakazaki, P. Echeverria.
Nation wide surveillance program to identify diarrhea-causing Escherichia coli in children in Thailand.
J Clin Microbiol, 28 (1990), pp. 469-472
[435]
M.T.G. Almeida, R.M. Silva, L.M. Donaire, L.E. Moreira, M.B. Martinez.
Enteropathogens associated with acute diarreal disease in children.
J Pediatr, 74 (1998), pp. 291-298
[436]
A.C. Moreno, A. Fernandes-Filho, T.A.T. Gomes, et al.
Etiology of childhood diarrhea in the northeast of Brazil: significant emergent diarrheal pathogens.
Diagn Microbiol Infect Dis, 66 (2010), pp. 50-57
[437]
D.M. Lozer, T.B. Souza, M.V. Monfardini, et al.
Genotypic and phenotypic analysis of diarrheagenic Escherichia coli strains isolated from Brazilian children living in low socioeconomic level communities.
BMC Infect Dis, 13 (2013), pp. 418
[438]
E.C. Souza, M.B. Martinez, C.R. Taddei, et al.
Etiology profile of acute diarrhea in children in São Paulo.
J Pediatr, 78 (2002), pp. 31-38
[439]
M.R.F. Toledo, L.R. Trabulsi.
Frequency of enteroinvasive Escherichia coli in children with diarrhea and healthy controls, in São Paulo, SP, Brazil.
Rev Microbiol, 21 (1990), pp. 1-4
[440]
M.G. Oliveira, G.V.A. Pessoa, L.K. Nakahara.
Enteropathogenic bacteria occurrence in diarrheic children living in Juiz de Fora municipality Minas Gerais Brazil.
Rev Inst Adolfo Lutz, 49 (1989), pp. 161-168
[441]
M.R.F. Toledo, L.R. Trabulsi.
Correlation between biochemical and serological characteristics of Escherichia coli and results of the Sereny Test.
J Clin Microbiol, 17 (1983), pp. 419-421
[442]
A. Ud-Din, S. Wahid.
Relationship among Shigella spp. and enteroinvasive Escherichia coli (EIEC) and their differentiation.
Braz J Microbiol, 45 (2014), pp. 1131-1138
[443]
F. Ørskov, I. Ørskov.
Escherichia coli serotyping and disease in man and animals.
Can J Microbiol, 38 (1992), pp. 699-704
[444]
B. Sereny.
Experimental Shigella keratoconjunctivitis. A preliminary report.
Acta Microbiol Acad Sci Hung, 2 (1955), pp. 293-296
[445]
E.V. Oaks, T.L. Hale, S.B. Formal.
Serum immune response to Shigella proteins antigens in rhesus monkeys and human infected with Shigella spp.
Infect Immun, 53 (1986), pp. 57-63
[446]
M. Pavlovic, A. Luze, R. Konrad, et al.
Development of a duplex real-time PCR for differentiation between E. coli and Shigella spp.
J Appl Microbiol, 110 (2011), pp. 1245-1251
[447]
M. Escher, G. Scavia, S. Morabito, et al.
A severe foodborne outbreak of diarrhoea linked to a canteen in Italy caused by enteroinvasive Escherichia coli, an uncommon agent.
Epidemiol Infect, 142 (2014), pp. 2559-2566
[448]
K. Sankaran, S. Banerjee, A.R. Pavankumar, M. Jesudason, R. Reissbrodt, P.H. Williams.
Apyrase-based colorimetric test for detection of Shigella and enteroinvasive Escherichia coli in stool.
Diagn Microbiol Infect Dis, 63 (2009), pp. 243-250
[449]
T. Bhargava, S. Datta, R. Ramachandran, R.K. Roy, K. Sankaran, Y.V.B.K. Subrahmanyam.
Virulent Shigella codes for a soluble apyrase: identification, characterization and cloning of the gene.
Curr Sci, 68 (1995), pp. 293-300
Copyright © 2016. Sociedade Brasileira de Microbiologia
Article options
Tools