17
Epidemiology and Clinical Manifestations of Enteroaggregative Escherichia coli Betina Hebbelstrup Jensen, a Katharina E. P. Olsen, a Carsten Struve, a Karen Angeliki Krogfelt, a Andreas Munk Petersen a,b,c Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmark a ; Department of Gastroenterology, Hvidovre Hospital, Copenhagen, Denmark b ; Department of Clinical Microbiology, Hvidovre Hospital, Copenhagen, Denmark c SUMMARY ..................................................................................................................................................614 INTRODUCTION ............................................................................................................................................614 PATHOGENICITY OF EAEC..................................................................................................................................615 Adherence................................................................................................................................................615 Biofilm Formation ........................................................................................................................................615 Toxins ....................................................................................................................................................615 Pathogenicity Islands .....................................................................................................................................616 Phylogeny of EAEC .......................................................................................................................................616 Volunteer Studies.........................................................................................................................................616 DIAGNOSIS OF EAEC .......................................................................................................................................617 CLINICAL MANIFESTATIONS ...............................................................................................................................617 Inflammatory Response ..................................................................................................................................617 Host Factors Are Determinants of EAEC Pathogenesis ...................................................................................................620 Immunological Response to EAEC Infection .............................................................................................................620 MULTIDRUG RESISTANCE IN EAEC STRAINS ...............................................................................................................620 RESERVOIR AND TRANSMISSION OF EAEC ................................................................................................................620 EAEC AS A CAUSE OF CHILDHOOD DIARRHEA IN DEVELOPING COUNTRIES ............................................................................621 ASSOCIATION BETWEEN EAEC AND HIV-RELATED DIARRHEA ............................................................................................621 TRAVELER’S DIARRHEA AND EAEC.........................................................................................................................622 ASSOCIATION BETWEEN EAEC AND DIARRHEA IN INDUSTRIALIZED COUNTRIES .......................................................................622 URINARY TRACT INFECTIONS AND EAEC ..................................................................................................................623 OUTBREAKS ................................................................................................................................................623 The German O104:H4 EAEC Outbreak in 2011............................................................................................................624 DISCUSSION ................................................................................................................................................624 CONCLUSIONS .............................................................................................................................................625 ACKNOWLEDGMENTS......................................................................................................................................625 REFERENCES ................................................................................................................................................625 AUTHOR BIOS ..............................................................................................................................................630 SUMMARY Enteroaggregative Escherichia coli (EAEC) represents a heteroge- neous group of E. coli strains. The pathogenicity and clinical rele- vance of these bacteria are still controversial. In this review, we describe the clinical significance of EAEC regarding patterns of infection in humans, transmission, reservoirs, and symptoms. Manifestations associated with EAEC infection include watery di- arrhea, mucoid diarrhea, low-grade fever, nausea, tenesmus, and borborygmi. In early studies, EAEC was considered to be an op- portunistic pathogen associated with diarrhea in HIV patients and in malnourished children in developing countries. In recent stud- ies, associations with traveler’s diarrhea, the occurrence of diar- rhea cases in industrialized countries, and outbreaks of diarrhea in Europe and Asia have been reported. In the spring of 2011, a large outbreak of hemolytic-uremic syndrome (HUS) and hemorrhagic colitis occurred in Germany due to an EAEC O104:H4 strain, causing 54 deaths and 855 cases of HUS. This strain produces the potent Shiga toxin along with the aggregative fimbriae. An outbreak of urinary tract infection associated with EAEC in Co- penhagen, Denmark, occurred in 1991; this involved extensive production of biofilm, an important characteristic of the patho- genicity of EAEC. However, the heterogeneity of EAEC continues to complicate diagnostics and also our understanding of pathoge- nicity. INTRODUCTION D iarrhea is still an important disease burden worldwide. It causes considerable childhood mortality in the developing world and is associated with morbidity and substantial health care costs in industrialized countries (1). One important cause of in- fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined: enterotoxigenic E. coli (ETEC), Shiga toxin-producing E. coli (STEC), enteroinvasive E. coli (EIEC), enteropathogenic E. coli (EPEC), and enteroaggregative E. coli (EAEC) (3–5). Recently, the pathogenicity of these pathotypes, including EAEC, was re- viewed (5); in this review, we focus on the epidemiology and dis- ease manifestations of EAEC specifically. EAEC was first described in 1987 by Nataro et al. in a study examining different patterns of Address correspondence to Karen Angeliki Krogfelt, [email protected]. Copyright © 2014, American Society for Microbiology. All Rights Reserved. doi:10.1128/CMR.00112-13 614 cmr.asm.org Clinical Microbiology Reviews p. 614 – 630 July 2014 Volume 27 Number 3 on May 20, 2021 by guest http://cmr.asm.org/ Downloaded from

Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

Epidemiology and Clinical Manifestations of EnteroaggregativeEscherichia coli

Betina Hebbelstrup Jensen,a Katharina E. P. Olsen,a Carsten Struve,a Karen Angeliki Krogfelt,a Andreas Munk Petersena,b,c

Department of Microbiology and Infection Control, Statens Serum Institut, Copenhagen, Denmarka; Department of Gastroenterology, Hvidovre Hospital, Copenhagen,Denmarkb; Department of Clinical Microbiology, Hvidovre Hospital, Copenhagen, Denmarkc

SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .614INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .614PATHOGENICITY OF EAEC. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .615

Adherence. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .615Biofilm Formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .615Toxins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .615Pathogenicity Islands . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .616Phylogeny of EAEC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .616Volunteer Studies. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .616

DIAGNOSIS OF EAEC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .617CLINICAL MANIFESTATIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .617

Inflammatory Response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .617Host Factors Are Determinants of EAEC Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .620Immunological Response to EAEC Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .620

MULTIDRUG RESISTANCE IN EAEC STRAINS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .620RESERVOIR AND TRANSMISSION OF EAEC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .620EAEC AS A CAUSE OF CHILDHOOD DIARRHEA IN DEVELOPING COUNTRIES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .621ASSOCIATION BETWEEN EAEC AND HIV-RELATED DIARRHEA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .621TRAVELER’S DIARRHEA AND EAEC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .622ASSOCIATION BETWEEN EAEC AND DIARRHEA IN INDUSTRIALIZED COUNTRIES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .622URINARY TRACT INFECTIONS AND EAEC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .623OUTBREAKS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .623

The German O104:H4 EAEC Outbreak in 2011. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .624DISCUSSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .624CONCLUSIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .625ACKNOWLEDGMENTS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .625REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .625AUTHOR BIOS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .630

SUMMARY

Enteroaggregative Escherichia coli (EAEC) represents a heteroge-neous group of E. coli strains. The pathogenicity and clinical rele-vance of these bacteria are still controversial. In this review, wedescribe the clinical significance of EAEC regarding patterns ofinfection in humans, transmission, reservoirs, and symptoms.Manifestations associated with EAEC infection include watery di-arrhea, mucoid diarrhea, low-grade fever, nausea, tenesmus, andborborygmi. In early studies, EAEC was considered to be an op-portunistic pathogen associated with diarrhea in HIV patients andin malnourished children in developing countries. In recent stud-ies, associations with traveler’s diarrhea, the occurrence of diar-rhea cases in industrialized countries, and outbreaks of diarrhea inEurope and Asia have been reported. In the spring of 2011, a largeoutbreak of hemolytic-uremic syndrome (HUS) and hemorrhagiccolitis occurred in Germany due to an EAEC O104:H4 strain,causing 54 deaths and 855 cases of HUS. This strain producesthe potent Shiga toxin along with the aggregative fimbriae. Anoutbreak of urinary tract infection associated with EAEC in Co-penhagen, Denmark, occurred in 1991; this involved extensiveproduction of biofilm, an important characteristic of the patho-genicity of EAEC. However, the heterogeneity of EAEC continues

to complicate diagnostics and also our understanding of pathoge-nicity.

INTRODUCTION

Diarrhea is still an important disease burden worldwide. Itcauses considerable childhood mortality in the developing

world and is associated with morbidity and substantial health carecosts in industrialized countries (1). One important cause of in-fectious diarrhea is the so-called diarrheagenic Escherichia coli(DEC) group (1, 2). The following subgroups of DEC have beendefined: enterotoxigenic E. coli (ETEC), Shiga toxin-producing E.coli (STEC), enteroinvasive E. coli (EIEC), enteropathogenic E.coli (EPEC), and enteroaggregative E. coli (EAEC) (3–5). Recently,the pathogenicity of these pathotypes, including EAEC, was re-viewed (5); in this review, we focus on the epidemiology and dis-ease manifestations of EAEC specifically. EAEC was first describedin 1987 by Nataro et al. in a study examining different patterns of

Address correspondence to Karen Angeliki Krogfelt, [email protected].

Copyright © 2014, American Society for Microbiology. All Rights Reserved.

doi:10.1128/CMR.00112-13

614 cmr.asm.org Clinical Microbiology Reviews p. 614 – 630 July 2014 Volume 27 Number 3

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 2: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

adherence of E. coli strains to HEp-2 cells in culture. The strainswere collected from Chilean children with diarrhea, and the typi-cal “stacked-brick” pattern was observed (6), which continues todefine EAEC (Fig. 1). The aggregative adherence (AA) pattern isdefined as the binding of bacteria to epithelial cells in a stacked-brick manner. Although EAEC has been associated with diarrheain studies conducted in both developing and industrialized coun-tries, it has been difficult to determine the specific mechanisms ofEAEC pathogenicity, which has made assessments of the clinicalrelevance of this microorganism difficult. Here, we present theliterature on this subject, concentrating on the pathophysiology ofEAEC.

PATHOGENICITY OF EAEC

Regarding infection with any of the subtypes of EAEC strains, thefollowing stages of pathogenesis have been described: (i) initialadherence to the mucosal surface, (ii) biofilm formation, and (iii)induction of an inflammatory response and release of toxins (7–12) (Fig. 2). Knowledge of the stages of EAEC pathogenicity hasbeen obtained from studies of in vitro cell cultures, animal models,and patients infected with EAEC (10, 13–17).

Adherence

Adhesion to the intestinal epithelium is facilitated by fimbriae andis the first step in the bacterial colonization of the gut (10, 18, 19)(Table 1). The aggregative adherence fimbriae (AAF) have beenfound to include 4 major variants, with distinct structures of thepilin subunits (AAF/I to AAF/IV) (19–22). The fimbriae can splayout from the bacteria due to the surface protein dispersin (9, 23),which is encoded by the aap gene. Dispersin is believed toinduce changes in the electrostatic surface of the lipopolysac-charide layer of the bacteria. This makes this protein importantfor the adherence properties of EAEC (9). The fimbriae bind tocomponents of the extracellular matrix of intestinal epithelialcells, such as laminin, collagen IV, cytokeratin 8, and fibronec-tin (24, 25). The aggregative pattern of these bacterial strains isthought to emerge from binding to the epithelial cell surfaceand binding to adjacent EAEC bacteria (26). Autoaggregation

contributes to the AA pattern, where adherence to epithelialcells also involves interactions between bacteria (18). The plas-mid-borne aggR gene is another important gene for the patho-genesis and adherence properties of EAEC, where strains pos-sessing the aggR gene are known as “typical EAEC strains” (27).aggR is a transcriptional activator that promotes the expressionof both chromosomal and plasmid-encoded virulence factors,including AAF and dispersin (28).

Biofilm Formation

When the gut has been colonized with EAEC, secretion of exces-sive mucus has been described, which is followed by the formationof biofilm (10, 29, 30). Biofilm formation is an important patho-genicity trait of EAEC and is situated mainly in the colon; how-ever, formation of biofilm in the small intestine has also beenreported (10). Biofilm formation may be an important contribu-tory factor in persistent infection by allowing the bacteria to evadethe local immune system and by preventing the transport of anti-bacterial factors, including antibiotics (31, 32). Assays to quantifybiofilm formation have been suggested as a possible method ofscreening for pathogenic EAEC strains (8). In several studies, ex-pression of AAF has been shown to be essential for biofilm forma-tion by EAEC (20, 24, 33–35). Other factors involved in the for-mation of biofilm include the shf gene, which has proven to beimportant for solid-biofilm production in EAEC reference strain042 (36). The genes yafK and fis are also important for biofilmformation, probably due to their involvement in the regulation ofAAF expression (33). Furthermore, in epidemiological studies,the plasmid-borne aatA gene (37), encoding the dispersin trans-porter; the set1A gene (38); and the aggR gene (39) have beenassociated with biofilm formation.

Toxins

Once the biofilm has been established, further damage to the in-testinal epithelium has been described, which is caused by therelease of bacterial toxins. The secretion of toxins is thought toplay an important role in secretory diarrhea, which is a typicalclinical manifestation of EAEC infection (4, 9, 40). The cytotoxiceffects of EAEC involve the secretion of serine protease autotrans-porters of the Enterobacteriaceae (SPATEs) (41). The SPATEs con-

FIG 2 Stages of pathogenesis of EAEC. Numbers in circles show the pro-gression of EAEC pathogenesis. (1) Agglutination of planktonic EAEC bac-teria. (2) Adherence to the intestinal epithelium and colonization of thegut. (3) Formation of biofilm. (4) Release of bacterial toxins, inducingdamage to the epithelium and increased secretion. (5) Establishment ofadditional biofilm.

FIG 1 Characteristic stacked-brick pattern observed when EAEC is culturedon HEp-2 cells. This pattern is also known as a honeycomb formation, whichemerges due to bacterium-bacterium and bacterium-cell interactions. The re-sult is the aggregation of the bacteria in a stacked-brick manner. (Courtesy ofRie Jønsson, Department of Microbiology and Infection Control, Statens Se-rum Institut, Copenhagen, Denmark; reprinted with permission.)

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 615

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 3: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

stitute a large family of extracellular proteases secreted by Entero-bacteriaceae via the type V secretion system (28). The SPATE genescan be either chromosomal or plasmid borne, and they are orga-nized into 2 phylogenetically different classes: class I SPATEs arecytotoxic to epithelial cells and include proteins encoded by thepet, sigA, and sat genes, whereas class II SPATEs have more diverseeffects and include proteins encoded by the pic and Shigella extra-cellular protease (sepA) genes (42, 43). Pet (plasmid-encodedtoxin) cleaves spectrin in the epithelial cytoskeleton (44), resultingin the deformation and exfoliation of the cell, and is associatedwith mucoid stools (45). Sat (secreted autotransporter toxin) hasbeen shown to cause loosening of cellular tight junctions in kidneycells and vacuolation in both kidney cells and bladder cells (46).Pic (protein involved in intestinal colonization) is a mucinase thatinterferes with the integrity of the mucus membrane and inducesserum resistance and hemagglutination (30, 47). The astA geneencodes EAST1 (EAEC heat-stable enterotoxin), which is not aSPATE and which shares certain functional properties of the en-terotoxin (STa) secreted by ETEC (48). This toxin causes in-creased secretion of chloride and has been associated with secre-tory diarrhea (49, 50). However, EAST1 is not confined to EAECstrains, as it has also been detected in EPEC, ETEC, and EHECstrains (50). Furthermore, the astA gene cannot be detected in allEAEC strains (51–53). The ShET1 enterotoxin encoded by the setgene was first identified in Shigella flexneri and may be associatedwith increased fluid secretion (54). The toxin-induced damageobserved in the intestinal epithelium, associated with EAEC infec-tion, other than that mentioned above includes hemorrhagicnecrosis and shortening of villi, enlarged crypt openings, and for-mation of crypt abscesses (53, 55, 56).

Pathogenicity Islands

Different pathogenicity islands have been identified in EAECstrains. One genomic island is inserted at the tRNA pheU locusand encodes the aaiC-associated type VI secretion system, whichis regulated by the aggR gene (28). The Shigella species she patho-genicity island found in some EAEC strains encodes the SPATEsPic and ShET1 enterotoxin, thereby conferring toxic and mucin-olytic activities (47). Two pathogenicity islands associated withextraintestinal E. coli strains, the Yersinia high-pathogenicity is-land, encoding the yersiniabactin siderophore, and the hly patho-

genicity island, encoding hemolysin and P-fimbriae, have alsobeen found in EAEC isolates (57).

Phylogeny of EAEC

Phylogenetic analysis of E. coli pathotypes segregates the strainsinto 6 major groups, groups A, B1, B2, C, D, and E (58–61).Extraintestinal E. coli strains belong largely to groups B2 and D,whereas commensal E. coli strains frequently belong to group A (5,61). One study investigated the phylogeny of 67 diarrheagenic E.coli strains, including 10 EAEC strains and 31 commensal E. colistrains. EHEC, Shigella species, and ETEC were found to belong togroups A, B1, C, and E, whereas EAEC strains were found to bescattered among all phylogenetic groups investigated (60). An-other study investigating EAEC phylogeny by multilocus enzymeelectrophoresis (MLEE) found 2 clusters of DEC containingEAEC strains. However, other pathotypes were found to fall intothe same clusters on the phylogenetic map (59). One epidemio-logical study investigated the potential clustering of EAEC strainsinto different phylogenetic groups but found strains in phylo-groups A, B1, B2, and D, and the authors of this study concludedthat EAEC originates from multiple lineages (62). The above-de-scribed studies indicate that EAEC strains are phylogeneticallydiverse and do not belong to specific phylogroups.

Volunteer Studies

EAEC as a gastrointestinal pathogen was investigated in a volun-teer study performed by Nataro et al. in 1995 (63). To identifypathogenic EAEC isolates, 4 groups of 5 volunteers were each fed1 of 4 different EAEC strains, 042, JM221, 17-2, or 34b, each at adose of 1010 CFU/ml. The strains were collected from patientssuffering from diarrhea in Peru, Mexico, Chile, and India, respec-tively. EAEC serotype O44:H18 strain 042 expressed AAF/II fim-briae, while strains 17-2, 34b, and JM221 expressed AAF/I fim-briae, which were identified by immunogold electron microscopyand DNA hybridization. EAEC strains 042 and 17-2 were found toexpress the gene encoding the enterotoxin EAST1. Strains JM221and 34b did not express EAST1. EAEC strain 042 caused diarrheain 3 of 5 adults; the 3 other EAEC strains failed to cause diarrhea.An earlier volunteer study (64) found that EAEC serotype O78:H33/35 strain JM221 at a dose of 7 � 108 CFU/ml was associatedwith diarrhea in 2 out of 8 volunteers and at a dose of 1 � 1010

TABLE 1 Key EAEC virulence genesa

Virulence gene(s) Function Location Role in pathogenesis

aggR Transcriptional activator of virulence genes Plasmid AdherenceAAF/I–AAF/IV genes Aggregative adherence fimbriae (4 variants) Plasmid Adherenceaap Antiaggregation protein dispersin Plasmid AdherenceaatA Dispersin transporter protein Plasmid Biofilm productionfis Regulation of AAF expression Chromosome Biofilm productionshf Encoding a Shigella flexneri homolog protein Plasmid Biofilm productionyafK Regulation of AAF expression Chromosome Biofilm productionastA Heat-stable enterotoxin EAST1 Plasmid Toxinspet Plasmid-encoded toxin Plasmid ToxinssepA Shigella extracellular protein Plasmid Toxinssat Secreted autotransporter toxin Chromosome Toxinsset Shigella enterotoxin 1 Chromosome Toxinspic Protein involved in colonization Chromosome Toxinsa The EAEC virulence genes listed are involved in several stages of pathogenicity. The appointed stage of pathogenicity for each virulence gene is given for simplicity.

Hebbelstrup Jensen et al.

616 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 4: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

CFU/ml was associated with diarrhea in 3 out of 8 volunteers. Theincubation period was shorter with the higher inoculum. The lackof consistency in the pathogenic assessment of the EAEC strains inthe 2 studies may have been be caused by the relatively small studygroups. The numbers of volunteers challenged with strain JM221differed between the studies, with 8 healthy adults being chal-lenged in the study by Mathewson et al. (64) and only 5 volunteersin being challenged in the study performed by Nataro et al. (63).The small numbers of volunteers challenged could have resultedin a misrepresentation of different host factors important forEAEC-induced disease in the two panels of volunteers. As revealedin later studies, host susceptibility is suspected to play a pivotalrole in EAEC pathogenesis, which could partly explain the differ-ent conclusions drawn from the volunteer studies.

DIAGNOSIS OF EAEC

The gold standard for the identification of EAEC remains theHEp-2 cell assay (6, 8, 31, 65). This test is performed only inreference laboratories; it requires cell culture facilities and is time-consuming (66). Molecular techniques have been developed todetect EAEC, of which PCR amplification of specific EAEC viru-lence-associated genes is of great importance. However, the ge-netic heterogeneity of EAEC and the notion that EAEC virulenceprobably results from a combination of multiple factors make it achallenge to differentiate pathogenic and nonpathogenic strainsby the molecular methods used today (31, 51, 67, 68). Variousgene targets have been used to detect EAEC by using PCR (Tables2 and 3). One EAEC-specific gene is the chromosomal aaiC gene,which is a gene in a genomic island encoding a type VI secretionsystem (28). Additional EAEC genes that are frequently used todetect EAEC by PCR include aggR and aatA (11, 65, 69–71).

In the volunteer study by Nataro et al. (63), EAEC strain 042,eliciting the stacked-brick pattern, was shown to cause diarrhea inthe majority of study participants, and since then, it has served asthe main EAEC reference strain. Reference strains representingspecific EAEC genes, for performing PCR and observing the ag-gregative pattern in the HEp-2 cell assay, include 042 (aatA, aggR,aap, and aaiC), JM221 (aggA and sat), 17-2 (aatA and aggR),55989 (agg3A and agg3/4C), and C1010 (agg4A, agg3/4C, sat, andsepA). AAF/I, -II, -III, and -IV are expressed by reference strains17-2, 042, 55989, and C1010, respectively (6, 22, 42, 66, 71, 72)(Table 2).

Serotyping of EAEC strains has proven to be unsuitable due tothe considerable diversity among strains and the large number ofso-called “rough” strains, which are strains that do not express anO antigen (73–76). Autoagglutination caused by the aggregative

phenotype is another obstacle when serotyping EAEC, leading to alarge number of nontypeable strains (23, 26, 42, 77). Serotypingand phylogenetic typing of EAEC have therefore proven to beuseful only in outbreak-related cases (78–82). The typical aggre-gative adherence pattern has been found to be associated with a60-MDa plasmid (pAA) (83). This plasmid carries several viru-lence factors, including the aggregative adherence fimbriae andtoxins (20). A DNA probe named CVD432 (also called the AAprobe) has been constructed from plasmid pAA for the detectionof EAEC by DNA hybridization (83). To identify EAEC, the spec-ificity of the CVD432 probe has been reported to be high but withvarious sensitivities compared to the HEp-2 cell assay (7, 52, 83,84). The lack of sensitivity comes from the genetic heterogeneityof the EAEC strains and the wide geographic dispersal of strainsanalyzed. As suggested in a study by Boisen et al. (20), the combi-nation of virulence genes may depend on the geographic region.International microbiological surveillance of EAEC and an im-proved understanding of EAEC pathogenesis could lead to theachievement of a proper diagnostic algorithm.

CLINICAL MANIFESTATIONS

The symptoms associated with EAEC infections include waterydiarrhea and occasionally very mucoid diarrhea (7, 29, 40), nau-sea, anorexia, low-grade fever, borborygmi, and tenesmus (39, 85,86). Cases of both acute and persistent diarrhea have been de-scribed. Persistent diarrhea is most frequently reported in chil-dren aged �1 year (32, 35, 87, 88). The site of colonization isbelieved to include the colon and the terminal ileum (10, 14, 55,89). The incubation time ranges from 8 h to 52 h (78, 90–92). Astudy by Steiner et al. in 1998 found that children in developingcountries who were diagnosed with EAEC infection suffered fromgrowth retardation regardless of the presence of diarrhea (93). Ina study by Roche et al. (13), growth retardation due to EAECinfection was observed in a mouse model. The growth impairmentwas found to be dependent on the dose of bacteria used for chal-lenge. It was observed that malnourished EAEC-inoculated micehad reduced growth velocity and increased shedding of EAEC instools compared to nourished mice. Bloody diarrhea has beenreported only rarely and involves mostly small children (10, 67,94, 95). However, the German O104:H4 EAEC Shiga toxin-ex-pressing outbreak strain caused hemorrhagic colitis and hemolytic-uremic syndrome (HUS), leading to considerable morbidity and ca-sualties (96–98). The outbreak strain contained the EAEC genes aggR,aggA, set1, pic, and aap and a prophage encoding the stx2 gene (99).Urinary tract infections (UTIs) associated with EAEC (46, 81, 100,101) and one case of urosepsis in an immunosuppressed female (102)have also been described recently. An outbreak of UTIs associatedwith EAEC in 1991 was reported in a Danish study (81), where theUTI outbreak strain contained the following combination of EAECgenes: sat, pic, aatA, aggR, aap, aaiC, and aggA.

Inflammatory Response

The inflammation caused by EAEC has been shown by increasedlevels of fecal interleukin-8 (IL-8), IL-1�, leukocytes, and lacto-ferrin, indicating a substantial gastrointestinal inflammatory re-sponse (11, 87, 103, 104). Occult blood has been detected in stoolsamples from EAEC-positive HIV patients (94). A study bySteiner et al. investigated childhood diarrhea and found increasedlevels of inflammation mediators, even in asymptomatic EAEC-positive carriers, compared to healthy EAEC-negative controls

TABLE 2 EAEC reference strainsa

Referencestrain Gene targets Serotype Reference

042 aatA, aggR, aap, aaiC O44:H18 6JM221 aggA, sat O92:H33 6417-2 aatA, aggR O3:H2 6C-1010 agg4A, agg3/4C, sat, sepA O?:H1 7255989 agg3A, agg3/4C O104:H4 22a Genes, according to the protein encoded, are as follows: aatA, outer membraneprotein; aggR, transcription activator; aap, antiaggregation protein dispersin; aaiC, partof a type VI secretion system; aggA, the major pilin subunit of AAF/I; agg3A, subunit ofAAF/III, sat, secreted autotransporter toxin; agg4A, subunit of AAF/IV; agg3/4C, usher-encoding region from AAF/III and AAF/IV; and sepA, Shigella extracellular protease.

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 617

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 5: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

TA

BLE

3G

eogr

aph

icdi

stri

buti

onof

EA

EC

case

sa

Ref

eren

ce(s

)Y

rN

o.of

pati

ents

(%E

AE

Cpo

siti

ve)

Cou

ntr

y(ie

s)V

iru

len

cefa

ctor

(s)

and/

ordi

agn

osti

cm

eth

od(s

)St

udy

type

Con

clu

sion

(s)

Val

idit

y

119

1997

798

case

s(2

),58

0co

ntr

ols

(0)

Ger

man

yP

rim

ers

com

plem

enta

ryto

the

CV

D43

2pr

obe

and

HE

p-2

cell

assa

y

Pro

spec

tive

case

-con

trol

EA

EC

was

asso

ciat

edw

ith

wat

ery

diar

rhea

and

chro

nic

diar

rhea

inch

ildre

nSt

ren

gth

sw

ere

larg

est

udy

grou

psan

ddi

arrh

eaco

nfi

rmed

bytr

ain

edpe

rson

nel

;wea

knes

ses

wer

eth

atE

AE

C-

posi

tive

pati

ents

wer

ete

sted

only

for

rota

viru

san

dth

atth

ede

fin

itio

nof

diar

rhea

was

only

2lo

ose

stoo

lsa

day

9319

9818

6(1

4)B

razi

lH

Ep-

2ce

llas

say

and

AA

prob

eC

ohor

tst

udy

EA

EC

-pos

itiv

ech

ildre

nh

adin

crea

sed

leve

lsof

lact

ofer

rin

and

IL-1

�in

stoo

lsam

ples

(P�

0.01

7)an

dsi

gnifi

can

tgr

owth

impa

irm

ent

rega

rdle

ssof

the

pres

ence

ofdi

arrh

ea(P

�0.

05)

Stre

ngt

hs

wer

eth

atit

was

lon

gitu

din

alst

udy

wh

ere

the

nu

trit

ion

alst

atu

san

dgr

owth

rate

sco

uld

beob

serv

edov

erti

me

and

that

stoo

lsam

ples

wer

ete

sted

for

para

site

san

dvi

ruse

s;w

eakn

esse

sw

ere

that

only

1E

.co

liis

olat

ew

asan

alyz

edfu

rth

erpe

rst

ools

ampl

ean

dth

atn

oda

taon

poss

ible

coin

fect

ion

sw

ere

avai

labl

e11

2002

176

case

s(4

9),1

0co

ntr

ols

(0)

Jam

aica

,In

dia,

and

Mex

ico

aggR

,aaf

A,a

ggA

,asp

U,

and

HE

p-2

cell

assa

yD

escr

ipti

ve/c

ase-

con

trol

aggR

-an

daa

fA-p

osit

ive

isol

ates

wer

eas

soci

ated

wit

hin

crea

sed

IL-8

leve

ls(P

�0.

05);

no

asso

ciat

ion

betw

een

AA

F/II

and

diar

rhea

was

obse

rved

Stre

ngt

hs

wer

eth

eu

seof

10h

ealt

hy

con

trol

str

avel

ing

toM

exic

oan

dth

atas

ympt

omat

icE

AE

Cca

rrie

rsw

ere

exam

ined

;wea

knes

ses

wer

eth

atn

oen

teri

cvi

ruse

sw

ere

test

edfo

ran

dth

atth

ere

was

no

men

tion

ofth

epo

ssib

leu

seof

anti

biot

ics

befo

rem

icro

biol

ogic

alte

stin

g69

2005

Neo

nat

es(n

oda

taon

nu

mbe

rs)

Sou

thK

orea

aggA

,aaf

Aag

gR,a

stA

,pic

,pe

t,C

VD

432

prob

e,an

dH

eLa

cell

assa

y

Scre

enin

gSo

me

EA

EC

prob

e-po

siti

veis

olat

esw

ere

not

posi

tive

inth

eH

Ep-

2ce

llas

say;

astA

and

aggR

wer

eth

em

ost

prev

alen

tE

AE

Cvi

rule

nce

gen

es

Stre

ngt

hs

wer

eth

orou

ghm

icro

bial

test

ing

usi

ng

HeL

ace

llas

say,

PFG

E,s

ensi

bilit

yte

stin

g,an

dD

NA

hyb

ridi

zati

on;w

eakn

esse

sw

ere

that

ther

ew

asn

oca

sede

fin

itio

nbe

side

sdi

arrh

eaan

dE

AE

Cst

atu

s,th

ere

was

no

info

rmat

ion

rega

rdin

gth

eu

seof

anti

biot

ics,

stoo

lsa

mpl

esw

ere

not

test

edfo

rga

stro

inte

stin

alvi

ruse

s,th

en

o.of

new

born

ste

sted

was

not

men

tion

ed,a

nd

only

the

no.

ofE

.col

iiso

late

ste

sted

was

men

tion

ed32

2007

122

case

s(5

5),1

27co

ntr

ols

(32)

Bra

zil

aggA

,aaf

Api

c,pe

t,as

tA,

CV

D43

2pr

obe,

and

HeL

ace

llas

say

Cas

e-co

ntr

olT

ypic

alE

AE

Cst

rain

sw

ere

fou

nd

ineq

ual

nu

mbe

rsin

both

grou

ps;t

he

astA

gen

ew

asas

soci

ated

wit

hac

ute

diar

rhea

(P�

0.01

);th

eC

VD

432

prob

ew

asas

soci

ated

wit

hpe

rsis

ten

tdi

arrh

ea(P

�0.

001)

Stre

ngt

hs

wer

eth

eex

clu

sion

ofpa

tien

tsw

ho

had

rece

ived

anti

biot

ics,

thos

ew

ho

had

con

gen

ital

gast

roin

test

inal

mal

form

atio

n,a

nd

thos

ew

ho

had

carb

ohyd

rate

into

lera

nce

and

mat

chin

gby

age

and

soci

oeco

nom

iccl

ass;

wea

knes

ses

wer

eth

atca

ses

wer

epo

orly

defi

ned

as“a

nin

crea

sein

evac

uat

ion

sw

ith

loos

est

ools

”an

dth

atth

ere

was

no

mic

robi

olog

ical

test

ing

for

gast

roin

test

inal

viru

ses

orpa

rasi

tes

inst

ools

ampl

es79

2007

7ca

ses

(71)

Japa

nas

tA,a

ggR

,CV

D43

2pr

obe,

and

HE

p-2

cell

assa

y

Ou

tbre

akA

nE

AE

Cst

rain

ofse

roty

peO

126:

H27

posi

tive

for

the

CV

D43

2pr

obe;

aggR

and

astA

wer

efo

un

din

4pa

tien

tsan

d1

food

han

dler

Stre

ngt

hs

wer

eth

atm

icro

biol

ogic

alte

stin

gin

clu

ded

PC

R,

PFG

E,H

Ep-

2ce

llas

say,

and

sero

typi

ng;

wea

knes

ses

wer

eth

atth

ere

was

no

test

ing

for

gast

roin

test

inal

viru

ses

orpa

rasi

tes

65,7

5,16

020

073,

506

case

s(5

),2,

772

con

trol

s(2

)U

Kaa

tA,a

ap,a

ggR

,agg

C,

agg3

C,a

afC

,shf

,pet

,pi

c,aa

iA,a

stA

,irp

2,ti

a,an

dH

Ep-

2ce

llas

say

Cas

e-co

ntr

olaa

fCw

asth

eon

lyge

ne

asso

ciat

edw

ith

diar

rhea

(P�

0.00

5);a

ggC

was

the

only

gen

eas

soci

ated

wit

hth

eco

ntr

olgr

oup

(P�

0.00

2);E

AE

Cw

asfo

un

dat

ah

igh

freq

uen

cyin

case

san

dco

ntr

ols

Stre

ngt

hs

wer

eth

atth

ere

was

aso

lidca

sede

fin

itio

nan

dre

duce

dbi

asby

excl

udi

ng

pati

ents

wit

hcy

stic

fibr

osis

,C

roh

n’s

dise

ase,

ulc

erat

ive

colit

is,o

rce

liac

dise

ase;

wea

knes

ses

wer

eth

atth

ere

was

no

info

rmat

ion

onpo

ssib

leco

infe

ctio

ns,

no

test

ing

for

gast

roin

test

inal

viru

ses

orpa

rasi

tes,

no

info

rmat

ion

from

stu

dygr

oups

rega

rdin

gtr

avel

,an

dn

oin

form

atio

non

dem

ogra

phic

s15

420

0925

3ca

ses

(7),

751

con

trol

s(3

)U

SAag

gR,a

ggA

,aaf

A,a

ap,

aatA

,ast

A,p

et,s

et1A

,an

dH

Ep-

2ce

llas

say

Cas

e-co

ntr

olst

udy

EA

EC

stra

ins

wit

hag

gR,a

atA

,ast

A,a

ap,a

nd

set1

Aw

ere

asso

ciat

edw

ith

diar

rhea

(P�

0.05

);ag

gR-p

osit

ive

stra

ins

cau

sed

hig

her

leve

lsof

IL-1

ra,I

L-6,

IL-8

,an

dtu

mor

nec

rosi

sfa

ctor

alph

a

Stre

ngt

hs

wer

eth

atth

ere

wer

ela

rge

stu

dygr

oups

,on

lypa

tien

tsw

ith

out

trav

elw

ith

in3

mo

wer

ein

clu

ded,

and

com

mu

nit

y-ac

quir

edE

AE

Cin

fect

ion

sco

uld

bede

tect

ed;w

eakn

esse

sin

clu

ded

ala

ckof

info

rmat

ion

onde

mog

raph

ics

inth

e2

grou

ps,p

osin

ga

poss

ible

sele

ctio

nbi

as78

2008

16ca

ses

(44)

Ital

yaa

p,as

tA,s

et1A

,aat

,agg

R,

and

HE

p-2

cell

assa

yO

utb

reak

An

EA

EC

stra

inw

ith

aggR

,aap

,an

dse

t1A

ofse

roty

peO

92:H

33w

asis

olat

edfr

om6

indi

vidu

als

and

1st

affm

embe

r;su

spec

ted

cau

seof

diar

rhea

was

un

past

euri

zed

chee

se

Stre

ngt

hs

wer

eth

atm

icro

biol

ogic

alte

stin

gof

food

sam

ples

and

stoo

lsam

ples

from

farm

anim

als

was

perf

orm

ed;w

eakn

esse

sw

ere

that

stoo

lsam

ples

wer

ete

sted

for

nor

ovir

us

only

and

not

for

oth

erga

stro

inte

stin

alvi

ruse

s

Hebbelstrup Jensen et al.

618 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 6: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

105

2010

170

case

s(8

7),1

04co

ntr

ols

(77)

Sou

ther

nG

han

aaa

tA,a

ap,a

ggR

,an

daa

iCP

rosp

ecti

vecr

oss-

sect

ion

alst

udy

EA

EC

was

asso

ciat

edw

ith

diar

rhea

(P�

0.04

8);E

AE

Cw

asas

soci

ated

wit

hin

crea

sed

feca

llac

tofe

rrin

leve

lsre

gard

less

ofth

epr

esen

ceof

diar

rhea

orn

utr

itio

nal

stat

us

Stre

ngt

hs

wer

eth

eu

seof

aqu

esti

onn

aire

onth

edu

rati

onof

diar

rhea

,dem

ogra

phic

s,liv

ing

con

diti

ons,

brea

stfe

edin

g,an

dco

nsu

mpt

ion

ofm

edic

ine

and

that

anth

ropo

met

ric

mea

sure

men

tsw

ere

incl

ude

din

the

stu

dy;w

eakn

esse

sw

ere

that

the

stoo

lsam

ples

wer

en

otte

sted

for

gast

roin

test

inal

viru

ses

133

2010

394

case

s(1

5),1

98co

ntr

ols

(24)

Sou

thIn

dia

aafI

I,aa

p,as

tA,a

ggR

,AA

prob

eC

ase-

con

trol

EA

EC

was

the

mos

tco

mm

only

isol

ated

DE

Cty

pein

both

grou

ps;t

he

com

bin

atio

nof

the

aap

and

aggR

gen

esan

dth

eA

Apr

obe

was

asso

ciat

edw

ith

diar

rhea

(P�

0.00

1)

Stre

ngt

hs

wer

eth

atch

ildre

nw

ere

test

edfo

rro

tavi

rus

and

Cry

ptos

pori

dium

spec

ies;

wea

knes

ses

wer

eth

atco

infe

ctio

ns

wer

em

enti

oned

,bu

tda

taon

the

dist

ribu

tion

inE

AE

C-p

osit

ive

pati

ents

wer

en

otgi

ven

5220

1014

0(1

1)Ir

anag

gR,a

afA

,agg

Aaa

p,as

tA,

prim

ers

com

plem

enta

ryto

the

CV

D43

2pr

obe,

and

HeL

ace

llas

say

Des

crip

tive

EA

EC

wit

hth

ege

net

icco

mbi

nat

ion

ofag

gR,

aap,

and

astA

was

the

mos

tpr

eval

ent;

only

86.7

%of

PC

R-i

den

tifi

edE

AE

Cba

cter

iaw

ere

posi

tive

inth

eH

eLa

cell

assa

y

Stre

ngt

hs

wer

eth

atin

form

atio

nre

gard

ing

sex,

age,

and

seas

onal

ity

was

avai

labl

e;w

eakn

esse

sw

ere

that

ther

ew

asn

oco

ntr

olgr

oup,

ther

ew

ere

no

data

avai

labl

eon

poss

ible

coin

fect

ion

s,an

dth

ere

was

no

info

rmat

ion

rega

rdin

gre

cen

ttr

avel

1720

1123

ulc

erat

ive

colit

is(6

1),

8C

roh

n’s

dise

ase

(88)

,23

con

trol

s(3

0)

Bra

zil

aggR

,pri

mer

sco

mpl

emen

tary

toth

eC

VD

432

prob

e,an

dH

Ep-

2ce

llas

say

Cas

e-co

ntr

olE

AE

Cw

asfr

equ

entl

yis

olat

edfr

ompa

tien

tssu

ffer

ing

from

Cro

hn

’sdi

seas

ean

du

lcer

ativ

eco

litis

wh

enex

amin

ing

E.c

oli

isol

ates

from

rect

albi

opsy

spec

imen

s

Stre

ngt

hs

wer

eth

ese

lect

ion

ofpa

tien

tsw

ith

wel

l-de

fin

edill

nes

ses

and

info

rmat

ion

onth

eu

seof

anti

biot

ics

and

age;

wea

knes

ses

wer

eth

atth

ere

was

no

test

ing

for

con

ven

tion

alen

tero

path

ogen

icba

cter

iaan

dn

ote

stin

gfo

rvi

ruse

sor

para

site

s15

720

1383

case

s(4

1),8

3co

ntr

ols

(41)

Bra

zil

aaiC

,aat

A,c

apU

,aap

,eilA

,ag

gR,o

rf3,

sat,

sepA

,pic

,si

gA,p

et,a

stA

,aaf

C,

agg3

/4C

,agg

3A,a

afA

,ag

gA,a

gg4A

,an

dor

f61

Cas

e-co

ntr

olpe

tor

aafA

was

sign

ifica

ntl

yas

soci

ated

wit

hdi

seas

e;th

eE

AE

Cst

rain

spo

sses

sin

gaa

iCan

dag

g3/4

Cbu

tla

ckin

gag

g4A

and

orf6

1w

ere

asso

ciat

edw

ith

diar

rhea

case

s

Stre

ngt

hs

wer

eth

ata

larg

epa

nel

ofE

AE

Cvi

rule

nce

gen

esw

assc

reen

edfo

rin

EA

EC

-pos

itiv

est

ools

ampl

es;

wea

knes

ses

wer

eth

atco

nve

nti

onal

ente

ropa

thog

enic

bact

eria

,vir

use

s,an

dpa

rasi

tes

wer

en

otte

sted

for

inst

ools

ampl

es

aA

wid

era

nge

ofge

ne

targ

ets

and

com

bin

atio

ns

ther

eofw

ere

use

dto

dete

ctE

AE

Cby

PC

R.H

eLa

cell

assa

ysca

nbe

use

dto

dete

ctth

esa

me

AA

patt

ern

asth

atde

tect

edby

the

HE

p-2

cell

assa

y.T

he

gen

es,a

ccor

din

gto

the

prot

ein

enco

ded,

are

asfo

llow

s:aa

iA,d

irec

tin

gse

cret

ion

ofaa

iC;a

aiC

,par

tof

aty

peV

Ise

cret

ion

syst

em;a

ggA

,maj

orpi

linsu

bun

itof

AA

F/I;

aafA

and

aafI

I,su

bun

itof

AA

F/II

;aaf

C,u

sher

,AA

F/II

I/IV

asse

mbl

yu

nit

;agg

3an

dag

g3A

,su

bun

itof

AA

F/II

I;ag

g4A

,su

bun

itof

AA

F/IV

;agg

C,u

sher

-en

codi

ng

regi

on;a

ggR

,tra

nsc

ript

ion

acti

vato

r;as

tA,a

ggre

gati

veh

eat-

stab

ileto

xin

1E

AST

1(e

nte

roag

greg

ativ

eE

.col

ihea

t-st

abile

toxi

n);

aap,

anti

aggr

egat

ion

prot

ein

disp

ersi

n;a

faD

,in

vasi

on;a

atA

,ou

ter

mem

bran

epr

otei

n;a

spU

,dis

pers

insu

rfac

eco

atpr

otei

n;c

apU

,hex

osyl

tran

sfer

ase

hom

olog

;cdt

,cyt

odet

ach

ing

toxi

n;e

ilA,E

AE

CH

ilAh

omol

og;f

yuA

,in

volv

edin

iron

sequ

estr

atio

n;h

da,a

dhes

in;h

ly,h

emol

ysin

;hr

a1an

d-2

,hea

t-re

sist

ant

aggl

uti

nin

s;ip

aH,i

nva

sion

;irp

2,Y

ersi

nia

bios

ynth

esis

gen

e;or

f3,c

rypt

icpr

otei

n;o

rf61

,pla

smid

-en

code

dh

emol

ysin

;pet

,pla

smid

enco

din

gto

xin

;pic

,pro

tein

invo

lved

inco

lon

izat

ion

;sat

,sec

rete

dau

totr

ansp

orte

rto

xin

;sep

A,S

hige

llaex

trac

ellu

lar

prot

ease

;Sh

ET

,set

,an

dse

t1A

,Shi

gella

ente

roto

xin

1;si

gA,S

hige

llaIg

A-l

ike

prot

ease

hom

olog

;shf

,cry

ptic

open

read

ing

fram

e;st

x,Sh

iga

toxi

n;a

nd

tia,

gen

epr

odu

ctpr

otei

nim

port

ant

for

inva

sion

.

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 619

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 7: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

(93). In another study performed in southern Ghana, childrenwho were asymptomatic carriers of EAEC showed increased levelsof fecal lactoferrin (105). These findings suggest that EAEC couldhave pathogenic potential in asymptomatic carriers. The IL-8 in-flammatory response has been shown to be partially caused byflagella (fliC) in a Caco-2 cell assay, as it was found that an aflagel-lar mutant of EAEC did not produce the same inflammatory re-sponse (106).

Host Factors Are Determinants of EAEC Pathogenesis

EAEC has been isolated in high numbers from stool samples ofasymptomatic carriers (35, 107–109), leading to the theory thatthe manifestation of gastrointestinal disease may depend on hostfactors (10, 91, 110). An American study investigated differentsingle nucleotide polymorphisms (SNPs) in the promoter regionof the CD14 gene as predictors of the development of traveler’sdiarrhea due to EAEC (111). The CD14 gene encodes a crucial stepin the inflammatory response to bacterial lipopolysaccharidestimulation by the innate immune system. This study found thatone SNP in the promoter region of the CD14 gene was associatedwith an increased risk of EAEC-induced diarrhea. Patients withthe CD14 �159 TT genotype were significantly associated withEAEC-induced diarrhea (P � 0.008) compared with healthy con-trols. The diverse clinical presentations reported for EAEC-posi-tive individuals support the hypothesis of a genetic predispositionfor the manifestation of illness. Other host factors that may beimportant for EAEC pathogenesis require further investigation.

Immunological Response to EAEC Infection

Dispersin has been shown to cause an immune response in trav-elers visiting Mexico. Five students with diarrhea were diagnosedas having EAEC in their stool samples, and they showed a signifi-cant increase in anti-dispersin IgG absorbance when serum sam-ples were compared before and after infection measured by anenzyme-linked immunosorbent assay (ELISA) (P � 0.01). Stu-dents with and those without diarrhea showed significantly higherlevels of IgG antibody to a recombinant dispersin protein uponreturning from Mexico than the levels measured in serum at base-line (P � 0.0001). However, students who tested positive for dis-persin antibodies at baseline were not protected from EAEC in-fection during their stay (85). In addition, dispersin has not beenfound to be restricted to EAEC strains, as it has also been detectedin nonpathogenic E. coli strains (112). Another study showed in-creased levels of IgA antibodies binding to crude EAEC antigens instool samples from 5 out of 10 American students returning fromGuadalajara, Mexico. In that study, EAEC strain JM221 was usedas the antigen in dot blot and Western blot analyses (113). A vol-unteer study also showed increased levels of antibodies to differentantigen preparations from EAEC strain JM221 measured in stoolsamples from patients who developed diarrhea after oral challengewith strain JM221. Five patients who did not develop diarrhea inthe trial period proved to have increased secretory IgA (sIgA) an-tibody levels in stool samples before challenge, suggesting possibleantibody protection (114). In a Brazilian case-control study, in-creased serum levels of IgM and IgG antibodies to the toxins Petand Pic were measured in children following an EAEC infection;however, antibodies to Pic were also observed in healthy children.No obvious correlation between EAEC strains carrying the pic andpet genes isolated from children and measured antibodies to Picand Pet were seen (77). The conclusion from these studies is that

none of the antibodies measured have proven to offer high speci-ficity for EAEC, which excludes their applicability in a clinicalsetting.

MULTIDRUG RESISTANCE IN EAEC STRAINS

A worrying high level of multidrug resistance among EAEC strainshas been reported in several studies (8, 52, 115, 116). In addition,extended-spectrum beta-lactamase (ESBL) production and in-creased resistance to quinolones in EAEC strains have been de-scribed (108, 115–117). A study conducted in southern India(118) investigated 64 EAEC strains and reported that 75% of thestrains were multidrug resistant, and resistance to ciprofloxacinwas found in 63.5% of the strains. The majority of the multidrug-resistant strains were identified in children �5 years old; this maybe caused by the higher prevalence of EAEC in this age group andby colonization rather than infection, leading to a long-term car-rier state, which may facilitate antibiotic pressure. This statementis supported by another study investigating ESBL production byEAEC strains, which was detected in 1.5% of children with diar-rhea and in 4.3% of children without diarrhea (determined byPCR targeting the CTX-M enzyme) (108). This study was per-formed in 2008 to 2009 in Nicaragua, and EAEC strains werefound to have levels of resistance to ampicillin and trimethoprim-sulfamethoxazole that were significantly higher than those ofEHEC and EPEC strains (P � 0.005) and to have higher levels ofresistance to amoxicillin than ETEC strains (P � 0.021).

In an Iranian study, multidrug resistance in 10 EAEC strainswas investigated, and it was found in 71.4% of strains, with re-duced resistance to ciprofloxacin being detected in 42.9% ofstrains (52); however, the small number of strains tested in thisstudy does not allow a generalized picture of resistance of EAECstrains. Another study investigated the presence of ESBL-produc-ing strains and detected ESBL in 5 of 51 EAEC strains isolatedfrom Spanish travelers who had visited India (116). The GermanEAEC O104:H4 outbreak strains showed ESBL production (99),and of Danish EAEC UTI O78:H10 outbreak strains, 37% werefound to be multidrug resistant (81). The conclusion from thestudies mentioned above is that although the proportion of ESBL-producing EAEC strains is small, it is still a cause for concern, andsuch strains are being increasingly reported. In the battle againstthe development of further antibiotic resistance, the use of fluidreplacement and supportive treatment is preferable for manage-ment of EAEC-induced diarrhea in uncomplicated cases. Empir-ical antimicrobial therapy of EAEC should be avoided when anti-biotic intervention is deemed necessary.

RESERVOIR AND TRANSMISSION OF EAEC

The reservoir for EAEC still has not been determined, but it isgenerally accepted to be human (97, 115, 119). The transmissionof EAEC is often described as being food borne or through con-taminated water, and as such, it is believed to be transmitted by thefecal-oral route (11). One study investigated the growth of EAECin drinking water (120), where bacteria were added to differentsources of bottled water. It was discovered that EAEC strainsmaintained their viability for up to 60 days at normal storagetemperatures. The survival of the bacteria was discovered to beprolonged in mineral water compared to spring water. This wassuggested to be due to the larger amounts of Ca2� and Mg2� inmineral water, inducing genetic competence. Surface water,which in some regions of Australia is a source of drinking water,

Hebbelstrup Jensen et al.

620 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 8: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

was investigated for the presence of pathogenic E. coli from vari-ous creeks, rivers, and drains before and after a storm in Brisbane,Australia. EAEC was identified by PCR targeting the aggR gene in36% of the water samples in the dry period and in 26% in the wetperiod and was found to be highly prevalent among the diarrhea-genic E. coli strains investigated (121). In a different study, watersamples from 46 aquatic locations in Bangladesh were collectedduring both the winter and summer seasons, and those investiga-tors found EAEC by PCR targeting a primer sequence comple-mentary to the CVD432 probe in 17% of the samples in the winterseason and 4% in the summer season (122). Another possibleroute of transmission of EAEC is food handling, which was inves-tigated in a study performed in Ouagadougou, Burkina Faso,where EAEC was detected by PCR targeting the pic and aggRgenes. EAEC was detected in 5 samples out of 120 samples ofdifferent meats and beef (123). Weaning foods as a possible routeof transmission of gastrointestinal pathogens was investigated in astudy performed in São Paolo, Brazil, and EAEC was found byDNA hybridization in 3 out of 100 milk samples from infant feed-ing bottles that were handled by mothers with low socioeconomicstatus (124). Animals as possible reservoirs of EAEC were investi-gated in a British study (125), and 1,227 E. coli isolates were inves-tigated as possible EAEC strains: 401 from cows, 406 from sheep,and 400 from pigs. However, no EAEC isolates were detected byusing the pAA probe. A different study investigated diarrheagenicE. coli strains from cats and dogs in Brazil and detected EAEC byPCR targeting the aggR gene. EAEC was detected in 7.4% of dogswith diarrhea, 3.9% of dogs without diarrhea, and one cat withoutdiarrhea (126). A French study investigated whether slaughter-house waste is a possible source of EAEC in rivers, but EAEC wasnot detected in wastewater or effluents by PCR targeting the aggR,aap, and aatA genes (127). EAEC has been isolated only rarelyfrom animal sources, and whether animals truly are potential res-ervoirs of EAEC is not conclusive. Whether companion animalsare accidental hosts of EAEC due to close contact with humans canonly be speculated upon. Sharing of E. coli strains among house-hold members and pets has been reported previously (128). Theabove-mentioned studies strongly indicate that one likely route oftransmission of EAEC is via contaminated food and water, butfurther research to exclude nonhuman reservoirs of EAEC shouldbe conducted.

EAEC AS A CAUSE OF CHILDHOOD DIARRHEA INDEVELOPING COUNTRIES

Early studies on the etiology of diarrhea in children living in de-veloping countries revealed that EAEC is highly prevalent (10, 93,129). A case-control study from 2004 (67) investigated the causeof diarrhea in Mongolian children. Here, EAEC was the most fre-quently detected type of DEC. Only EAEC strains with the aggRgene were found to be associated with diarrhea. A possible con-founder in the study may have been gastrointestinal viruses, forwhich no testing was performed. In 1999, a case-control study wasperformed in Kolkata, India, where childhood diarrhea was inves-tigated, and EAEC strains were detected in stool samples by aHeLa cell assay and PCR amplification of a gene product comple-mentary to the CVD432 probe. EAEC was found to be associatedwith diarrhea in children �36 months of age (130). In this study,watery diarrhea was reported more frequently than mucoid diar-rhea among EAEC-positive children (72% versus 28%). A studyby Steiner et al. in 1998 (93) found that growth impairment and

increased levels of fecal inflammation markers were associatedwith EAEC infection in children living in a Brazilian urban slum.EAEC was detected by the AA gene probe and the HEp-2 cell assay,and EAEC-positive children in this study had impaired growthregardless of clinical manifestations (Table 3). Another case-con-trol study investigating childhood diarrhea failed to detect anyassociation with EAEC, but there was a high rate of carriage ofEAEC among controls. This study was performed in Dhaka, Ban-gladesh, from 1993 to 1994, and EAEC was detected by DNAhybridization using the CVD432 probe (131, 132). A high rate ofcarriage of EAEC in young children has been reported by severalother studies. EAEC was found in greater numbers in the controlgroup in a case-control study (133) conducted between 2003 and2006 in South India. This study investigated the etiology of diar-rhea in children �5 years of age. EAEC was detected by multiplexPCR targeting the astA, aap, aggR, and aafII genes and by PCRamplification of a product corresponding to the AA probe. EAECwas found in 14.7% of cases and 23.7% of controls. However,EAEC strains carrying only the aap, aggR, and genes correspond-ing to the AA probe were significantly associated with diarrhea,with incidences of 6.1% in cases and 3.0% in controls (P � 0.001).Another recent study found a high rate of asymptomatic carriersof EAEC in Mali, where diarrhea in children aged 0 to 59 monthswas investigated. After 1 year of surveillance, EAEC was found tobe the only DEC in 60 children with diarrhea and in 61 childrenwithout diarrhea. However, EAEC strains with the sepA gene werefound to be associated with diarrhea (odds ratio [OR], 5.6; P �0.0006) (42). In conclusion, the many studies on childhood diar-rhea and EAEC indicate that there is a valid association, but anexhaustive diagnostic method to detect pathogenic EAEC strainsis required, and the study design should include simultaneoustesting for conventional gastrointestinal pathogens. In addition,to assess the pathogenicity of EAEC in childhood diarrhea in gen-eral, case-control studies should be conducted in a Western set-ting to minimize or eliminate contributing disease-related factorssuch as malnutrition and poor hygiene.

ASSOCIATION BETWEEN EAEC AND HIV-RELATED DIARRHEA

HIV-related diarrhea was linked to EAEC in early studies, andEAEC was considered an opportunistic pathogen. A Swiss studyconducted between 1996 and 1998 investigated HIV-related diar-rhea (134), where EAEC was detected by the HeLa cell assay, theCVD432 probe, and PCR primers complementary to the CVD432probe. In this study, a low CD4 lymphocyte count was not foundto be a predisposing factor for EAEC infection. Of the 7 patientswho experienced diarrhea, 4 later suffered from persistent diar-rhea or chronic intermittent diarrhea. Tests for enteric parasites,cytomegalovirus, and enteropathogenic bacteria were included inthe testing of the stools. A study conducted in Senegal (135) foundEAEC in 31 of 158 HIV patients with diarrhea and in 3 of 160 HIVpatients without diarrhea. EAEC was detected by PCR amplifica-tion of a product corresponding to the CVD432 probe (136). Inthat study, EAEC was defined as an opportunistic pathogen. Anassociation between EAEC and diarrhea has not been reported byall studies on HIV-related diarrhea. EAEC was found in equalnumbers in diarrhea cases and controls among HIV-positive chil-dren in Peru (137), where EAEC was detected by PCR targetingthe aggR gene. Another study found that HIV patients were morelikely to be EAEC positive: 29.5% versus 14% in the HIV-negativegroup. EAEC was detected by multiplex PCR targeting aggR and

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 621

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 9: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

aap, the AA probe, and quantitative real-time PCR targeting theaggR gene (94). EAEC was found to be associated with occultblood in stool samples, (OR, 5.096; confidence interval, 2.665 to9.644), and an association between bacterial load and the mani-festation of illness was reported.

EAEC continues to play an important role in detection of thecause of diarrhea in HIV patients. However, the lack of an associ-ation between diarrhea in EAEC-positive patients and CD4 lym-phocyte counts does not support the theory of opportunisticpathogenicity, and today, the association between EAEC and di-arrhea is not limited to this group of patients.

TRAVELER’S DIARRHEA AND EAEC

More recent studies have found an association between EAEC andtraveler’s diarrhea. Initially, this was found for patients returningfrom developing countries, but now, the association has been re-ported in cases of traveler’s diarrhea obtained from various desti-nations. One study of traveler’s diarrhea concluded that the EAECvirulence factors aggR and aggA were the most prevalent EAECgenes detected in European and American patients with diarrheareturning from Montego Bay, Jamaica; Goa, India; and Guadala-jara, Mexico (11). Another study found the combination of theaggR gene and the aat gene to be associated with traveler’s diar-rhea; this combination was detected in 45% of cases, as opposed to16.4% of controls (P � 0.01). One strength of this study was thatcases of traveler’s diarrhea were compared with travelers who didnot have diarrhea and who were returning from the same regionsin the tropics of Asia, Africa, and Latin America (138). An Amer-ican study (29) investigated cases of traveler’s diarrhea from India,Guatemala, and Mexico. Forty-nine travelers had diarrhea andEAEC, 15 had EAEC without diarrhea, and 17 neither had diar-rhea nor tested positive for EAEC. EAEC strains with the aggR,astA, set1A, and aap genes were associated with traveler’s diarrhea(P � 0.05). Coinfections were reported to occur at a high fre-quency in another study of patients returning from India, Ja-maica, and Mexico (139). The most frequent pathogen isolated inthat study was ETEC, which was found in 41 out of 72 mixedinfections with EAEC detected by the HEp-2 cell assay. The pres-ence of another well-established pathogen, i.e., coinfection, inthese patients’ stool samples complicates any association betweendiarrhea and EAEC. A high number of coinfections was reportedby a study investigating traveler’s diarrhea among patients whohad visited Benin, West Africa (140). This study found that 79% ofthe patients were coinfected, where EPEC and EAEC were themost frequently detected pathogens. Coinfections with EAECwere also mentioned in another study but were described only asbeing scattered among cases of traveler’s diarrhea. This study in-vestigated Korean patients returning from Southeast Asian coun-tries (141), where ETEC had the highest incidence of entericpathogens detected (found in 36.0% of cases), followed by EAEC(found in 27.0% of cases). EAEC was detected by PCR, but theprimers used in this study were not described. Other pathogensdetected were Vibrio parahaemolyticus, Vibrio cholerae, Salmonellaspecies, Shigella, and norovirus.

EAEC is often isolated in cases of traveler’s diarrhea from dif-ferent countries, but the strong association found may have beenoverestimated due to the vast number of studies on EAEC per-formed for this patient category. The large number of coinfectionsand the lack of exhaustive testing for enteric pathogens in thesepatients mean that the general assessment of the pathogenic con-

tribution of EAEC to cases of traveler’s diarrhea must still be re-garded as questionable.

ASSOCIATION BETWEEN EAEC AND DIARRHEA ININDUSTRIALIZED COUNTRIES

In studies of the etiology of diarrheal episodes in industrializedcountries, the prevalence of EAEC has been reported to be high. Astudy conducted in Germany in 1997 found EAEC to be the thirdmost commonly isolated microorganism in young children withdiarrhea (2%), after Salmonella species (13.4%) and STEC (3.1%).EAEC infection was diagnosed in 16 cases by using the HEp-2 cellassay and PCR primers complementary to the CVD432 probe.EAEC was not detected in an asymptomatic group of children(119). However, a prospective American study did not find anassociation between EAEC and childhood diarrhea in the commu-nity setting (142). This study involved 604 healthy 6- to 36-month-old children who were monitored over a 6-month period,and the incidence of diarrhea was reported to be 2.2 per person-year. Children with intestinal malabsorption, inflammatory boweldisease, or cystic fibrosis and those who were treated with antibi-otics were excluded. Testing for enteric viruses, parasites, and en-teropathogenic bacteria was performed. Whether or not the ex-clusion of children with the comorbidities mentioned above inthis study played an important part in the pathogenic potential ofEAEC can only be speculated upon but warrants further research.An investigation of possible host factors contributing to EAEC-induced disease in children in industrialized countries should beconducted. A case-control study performed in Cincinnati, OH,found that children �1 year of age in the case group had EAECisolated significantly more frequently from stool samples than didchildren in the control group. Interestingly, pathogenic EAECstrains were identified only by the CVD432 probe and not by theaggregative adherence pattern observed in the HEp-2 cell assay.Strains with the aggregative adherence phenotype were equallydistributed between groups (143). Aggregative adherence in theHEp-2 cell assay was observed in E. coli strains that did not possessany known EAEC genes in a British study investigating commu-nity-acquired diarrhea (73). The HEp-2 cell assay was reported todetect various EAEC genotypes. This study used PCR primers tar-geting the aat, aaiA, and astA genes. Stool samples were collectedas part of a routine examination for diarrhea by the patients’ gen-eral practitioner, which included testing for EPEC, Salmonellaspecies, Shigella sonnei, Shigella flexneri, Clostridium difficile, Cam-pylobacter species, enteric parasites, and viruses. EAEC strainswith a combination of the aat, aai, and/or astA gene were found in39 of 500 patients with diarrhea. Some EAEC-positive patientswere coinfected with, for example, Campylobacter species (3 cases)and rotavirus (1 case). Foreign travel was associated with only aminor proportion of the EAEC-positive cases.

EAEC was associated with diarrhea in a national surveillancestudy conducted from 1993 to 1996 and from 2008 to 2009 in theUnited Kingdom (144). It was shown that approximately 3.3% ofintestinal infectious diseases were associated with EAEC, and theburden of EAEC was measured by quantitative PCR amplificationof a product corresponding to the CVD432 probe. However, theEAEC burden determined by quantitative PCR was not useful fordiscrimination between EAEC strains isolated from positive casesand EAEC strains isolated from controls.

In conclusion, EAEC is frequently isolated in industrializedcountries, not only from outbreaks, and should be considered

Hebbelstrup Jensen et al.

622 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 10: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

for inclusion in a diagnostic algorithm for enteric pathogens inthese settings. However, more case-control studies should beperformed in Western countries to identify the possible patho-genic contribution of EAEC to diarrheal episodes in theseareas.

URINARY TRACT INFECTIONS AND EAEC

Recently, EAEC strains have been associated with urinary tractinfections (UTIs) (46, 100, 101, 145). An outbreak of UTI casesassociated with an EAEC strain also possessing extraintestinalpathogenic E. coli (ExPEC) genes was reported in 1991 in a Danishstudy (81). In that outbreak, 18 patients suffering from UTI in theCopenhagen area were found to be infected by strains of the sameserotype, O78:H10, and the same phylogenetic group (group A).The outbreak strain had the ExPEC genes fyuA, traT, and iutA andthe EAEC genes sat, pic, aap, aaiC, aatA, aggR, and aggA. In a laterstudy, which further characterized the Copenhagen outbreakstrains, it was shown that most of the 19 strains were capable ofbiofilm formation. One of the outbreak isolates, originating froma child aged 4 months with recurrent UTI, proved to form exten-sive biofilms and pronounced adhesion to human bladder epithe-lial cells by the expression of AAF/I (34). This indicates thatEAEC-associated virulence factors enhance uropathogenicity, andthe possession of these virulence factors may have promoted thecapability of the strain to cause the outbreak.

A case of urinary tract infection preceded by diarrhea caused byEAEC was reported in a 55-year-old immunocompromised fe-male, leading to bacteremia and urosepsis (102). In this study, anEAEC strain with the same pulsed-field gel electrophoresis(PFGE) profile was isolated from stool, urine, and blood culturesamples from this patient. The strain was serotyped as O176:NTand tested positive for the CVD432 probe and the aggR, aap,agg3C, astA, pic, aaiC, and air genes by PCR. In a Korean study(145), the cause of UTI in small children was investigated by ana-

lyzing suprapubic urine specimens from children with and thosewithout UTI (as defined by numbers of CFU in urine). EAEC wasdetected by PCR targeting the aggR and aap genes in children withcases of UTIs; however, strains with the combination of aggR andaap were found in significantly higher numbers in the controlgroup (P � 0.03). EAEC isolated from patients with UTI, withdifferent combinations of the EAEC genes aap, aggR, aggC, astA,agg3C, pet, and pic but with all strains containing genes encodingAAF/I, were reported in a Brazilian study (100). These authorssuggested that this fimbria might play an important role in theuropathogenesis of EAEC.

In general, characterization of E. coli isolated from UTI cases islimited in the clinical setting, which obscures the pathogenic con-tribution from different uropathogenic E. coli groups. However,true outbreaks of UTI cases are rare, implicating little knowledgeon this subject. Clearly, the role of EAEC in uropathogenesisshould be further investigated.

OUTBREAKS

Outbreaks of gastroenteritis linked to EAEC have been reported(Fig. 3). One outbreak took place in a Serbian nursery (80) in1995, where EAEC was detected by the HEp-2 cell assay and wascharacterized as belonging to serotype O4 in 12 of 19 babies whohad low-grade fever, diarrhea, and weight loss. Rotavirus was theonly gastrointestinal virus screened for. PCR was not performedon the outbreak strain. Another outbreak took place in a policeinstitute in Japan in 2005, where the staff experienced gastroen-teritis after having consumed food suspected of being contami-nated with EAEC (79). Four staff members and one food handlertested positive for EAEC in stool samples; the isolates were foundto have an identical serotype (O126:H27). However, no microbi-ological investigations for enteric viruses were done for this out-break. The largest reported outbreak of EAEC, apart from the2011 German O104:H4 EAEC outbreak, took place in the Gifu

FIG 3 Geographic distribution of EAEC outbreaks. N is the number of patients with confirmed EAEC involved in the outbreak. The total numbers of patientsinvolved in outbreaks of diarrhea are given in brackets. The Danish outbreak was an outbreak of EAEC causing urinary tract infections. References are shown inparentheses. HUS, hemolytic-uremic syndrome.

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 623

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 11: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

prefecture, Tajimi, Japan, in 1993 (90). Here, 2,697 children de-veloped signs of food poisoning after having consumed schoollunches. Fecal specimens were collected from 30 children withsevere protracted diarrhea, and 12 tested positive for EAEC by theHEp-2 cell assay and the astA gene by PCR; the strains were all Ountypeable:H10. However, the children in this outbreak were nottested for gastrointestinal viruses, to which the symptoms corre-spond, including short-term gastroenteritis, nausea, stomachache, and diarrhea, which affected a considerable number of thecases in the outbreak. Another EAEC outbreak took place in 2008in Italy at a farm holiday resort, where the guests experiencedgastroenteritis after having consumed unpasteurized cheese (78).EAEC was isolated in stool samples from 6 restaurant guests and 1staff member; the strains were identified by the HEp-2 cell assay.The EAEC strain involved was identified as belonging to serotypeO92:H33, and it tested positive for the virulence genes aggR, aat,aap, and set1A by PCR. Testing for norovirus, but not rotavirus,was performed in that study.

The outbreaks mentioned above show a considerable potentialfor food-borne transmission of EAEC. The lack of internationalsurveillance of EAEC could lead to missed outbreaks caused byEAEC.

The German O104:H4 EAEC Outbreak in 2011

In May 2011, a large food-borne outbreak of Shiga toxin-produc-ing E. coli (STEC) occurred in Germany, with 3,842 confirmedcases. The source of the food-borne outbreak was bean sprouts,which caused 855 (22%) cases of hemolytic-uremic syndrome andconsiderable mortality (amounting to 54 [1.4%] deaths) (146).The outbreak strain was found to show the characteristic stacked-brick pattern when grown on HEp-2 cells, and it tested positive forthe EAEC genes aggR, aggA, set1, pic, and aap (99). The outbreakstrain was serotyped as O104:H4 and contained a prophage en-coding the Shiga toxin 2 gene, stx2, which is normally absent inEAEC strains (98). Other, considerably smaller, occurrences ofEAEC-STEC hybrid stains have been reported (147–149). An out-break of HUS in 10 children in France in 1992 was associated withan EAEC strain of serotype O111:H2, which was positive for thestx2 gene (150). A 3-year-old boy from Japan was diagnosed withHUS in 1999, where an EAEC strain of serotype O86:HNM whichwas positive for the stx2 gene was identified (148). An EAEC O111:H21 strain with the stx2 gene was found to be associated with anoutbreak of HUS in a household in Ireland in 2012 (149). AnEAEC strain of serotype O104:H4, but lacking the Shiga toxin, waspreviously detected in an HIV-positive patient in the Central Af-rican Republic who was suffering from persistent diarrhea (22).Further characterization of the outbreak strain by whole-genomephylogenetic analysis showed a cluster of genes with a close geneticrelationship between the African non-Shiga toxin-positiveO104:H4 strain and the outbreak strain (98), which suggested arecent acquisition of the phage carrying stx2. The outbreaks men-tioned above show the considerable potential of EAEC to acquireadditional virulence through genetic exchange, and this furtheremphasizes the need for microbiological surveillance.

DISCUSSION

The CVD432 probe targeting the aat gene is widely used for thedetection of EAEC by DNA hybridization (17, 52, 69, 79, 119,131). However, this probe fails to detect all EAEC strains showingthe stacked-brick pattern by the HEp-2 cell assay (65, 69, 106). The

possibility of interobserver variability in pattern recognition wasmentioned in a study by Pabst et al. (151), which could lead todifferent rates of detection of EAEC between studies. Indeed,strain JM221 was first described to show localized adherence (64)but has since been said to show aggregative adherence (63). Onecase-control study found patterns of adherence of EAEC strains(found by PCR testing for the aatA and aggR genes) to be diverse,including nonadherent, sparsely adherent, and densely adherentpatterns, without any correlation with cases or controls (95).

PCR is a less demanding and more objective method for thedetection of pathogenic E. coli. Extensive studies have been per-formed by using PCR to find the right combination of genes iden-tifying the “truly pathogenic” EAEC strains (51, 65, 69, 79, 105,152, 153); however, no consensus has been reached on this matter.As suggested by another study (20), the combination of EAECgenes might depend on the geographic region, and recognitionof these various combinations of genes could lead to a betterunderstanding of their role in the pathogenicity of specificEAEC strains.

The aggR gene is highly conserved among EAEC strains and hasbeen found to be associated with diarrhea in several studies (105,133, 154). A strong correlation between the CVD432 probe andthe aggR gene has been reported (7, 52, 69). Quantitative PCRtargeting the aggR gene has shown dose dependency (94) in themanifestation of illness. However, several other studies have notbeen able to associate aggR with disease (42, 65, 74, 155). Anotherway of quantifying the EAEC burden was used in a study usingDNA extraction from stools, which showed a log-linear correla-tion between CFU of E. coli per gram of stool and median fluores-cence intensity determined by multiplex PCR-Luminex technol-ogy. A correlation between the aatA gene and CFU of EAEC instool was found. Further research on this method, combined withclinical information, could reveal a possible correlation betweenthe degree of illness and the EAEC burden (156).

Persistent diarrhea has frequently been mentioned in thestudies reviewed, but it appears to lack solid documentation.Several studies have found a low prevalence of persistent diar-rhea in EAEC-positive patients (86, 129, 151, 157), which callsinto question the importance of this disease trait in an EAECinfection in general. Despite the genetic heterogeneity of EAECstrains, the reported clinical manifestations have a high level ofhomogeneity (8, 86, 134, 158, 159), with symptoms such aslow-grade fever, watery or mucoid diarrhea, borborygmi, andloss of appetite.

In a number of studies, EAEC has been isolated in almostequal numbers from patients suffering from diarrhea and fromcontrol groups (15, 133, 137). This finding has been proposedto be a result of acquired immunity (105, 137) due to poorsanitary conditions as well as frequent exposure to pathogenicE. coli strains, resulting in the development of protective antibod-ies and colonization rather than infection. This has been stated instudies conducted in developing countries. However, high num-bers of asymptomatic carriers of EAEC have also been observed incountries such as the United States, Switzerland, and Denmark(40, 72, 109, 142, 143). Case-control studies performed in settingswith poor hygiene are difficult to interpret in terms of key EAECvirulence genes, due to the high prevalence of EAEC in these areas.The reported long-term carrier state and the low pathogenicity ofEAEC might lead to colonization rather than infection, whichcould lead to incorrect conclusions about truly pathogenic EAEC

Hebbelstrup Jensen et al.

624 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 12: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

virulence genes when EAEC strains are compared between casesand controls. As was mentioned previously (95), the case-controlstudy design performed in a community with a high carrier rate ofEAEC complicates the assessment of pathogenicity.

Coinfections, especially in traveler’s diarrhea, are said to occurfrequently (138); however, few studies have investigated the etiol-ogy of diarrhea, including testing for DEC, viruses, and parasites,thus leaving the pathogenicity of EAEC difficult to assess in thesecases. In addition, possible synergy between these microorgan-isms should be investigated.

CONCLUSIONS

EAEC has been isolated from many patients suffering from diar-rhea in a number of regions of the world and from various socio-economic strata. It is most frequently reported as a self-limitingdiarrheagenic pathogen that generally results in mild symptoms.The importance of host factors as determinants of EAEC patho-genesis should be investigated in depth. Persistent diarrhea causedby EAEC is often mentioned in studies, but further research onthis matter is needed. The genetic heterogeneity of EAEC compli-cates international surveillance and therapeutic approaches con-siderably. The lack of a specific diagnostic algorithm to detectEAEC makes our understanding of the pathogenicity of EAECespecially difficult.

ACKNOWLEDGMENTS

We thank Rie Jønsson, Department of Microbiology and Infection Con-trol, Statens Serum Institut, for inspiring discussions and Fig. 1. We alsothank Marian Jørgensen, Department of Microbiological Diagnostics andVirology, Statens Serum Institut, for critical comments. We are grateful toAnja Bjarnum for assistance with graphical illustrations.

Funding was provided by Danish Council for Strategic Research, In-novation and Higher Education grant number 2101-07-0023 to KarenAngeliki Krogfelt.

We declare that there are no conflicts of interest.

REFERENCES1. Farthing M, Salam MA, Lindberg G, Dite P, Khalif I, Salazar-Lindo E,

Ramakrishna BS, Goh KL, Thomson A, Khan AG, Krabshuis J, LemairA. 2013. Acute diarrhea in adults and children: a global perspective. J.Clin. Gastroenterol. 47:12–20. http://dx.doi.org/10.1097/MCG.0b013e31826df662.

2. Kotloff KL, Blackwelder WC, Nasrin D, Nataro JP, Farag TH, van EijkA, Adegbola RA, Alonso PL, Breiman RF, Faruque AS, Saha D, SowSO, Sur D, Zaidi AK, Biswas K, Panchalingam S, Clemens JD, CohenD, Glass RI, Mintz ED, Sommerfelt H, Levine MM. 2012. The GlobalEnteric Multicenter Study (GEMS) of diarrheal disease in infants andyoung children in developing countries: epidemiologic and clinicalmethods of the case/control study. Clin. Infect. Dis. 55(Suppl 4):S232–S245. http://dx.doi.org/10.1093/cid/cis753.

3. Nataro JP, Kaper JB. 1998. Diarrheagenic Escherichia coli. Clin. Micro-biol. Rev. 11:142–201.

4. Arenas-Hernández MM, Martínez-Laguna Y, Torres AG. 2012. Clini-cal implications of enteroadherent Escherichia coli. Curr. Gastroenterol.Rep. 14:386 –394. http://dx.doi.org/10.1007/s11894-012-0277-1.

5. Croxen MA, Law RJ, Scholz R, Keeney KM, Wlodarska M, Finlay BB.2013. Recent advances in understanding enteric pathogenic Escherichiacoli. Clin. Microbiol. Rev. 26:822– 880. http://dx.doi.org/10.1128/CMR.00022-13.

6. Nataro JP, Kaper JB, Robins-Browne R, Prado V, Vial P, Levine MM.1987. Patterns of adherence of diarrheagenic Escherichia coli to HEp-2cells. Pediatr. Infect. Dis. J. 6:829 – 831. http://dx.doi.org/10.1097/00006454-198709000-00008.

7. Okeke IN, Macfarlane-Smith LR, Fletcher JN, Snelling AM. 2011. IS3profiling identifies the enterohaemorrhagic Escherichia coli O-island 62

in a distinct enteroaggregative E. coli lineage. Gut Pathog. 3:4. http://dx.doi.org/10.1186/1757-4749-3-4.

8. Bangar R, Mamatha B. 2008. Identification of enteroaggregative Esch-erichia coli in infants with acute diarrhea based on biofilm production inManipal, South India. Indian J. Med. Sci. 62:8 –12. http://dx.doi.org/10.4103/0019-5359.38916.

9. Harrington SM, Dudley EG, Nataro JP. 2006. Pathogenesis of entero-aggregative Escherichia coli infection. FEMS Microbiol. Lett. 254:12–18.http://dx.doi.org/10.1111/j.1574-6968.2005.00005.x.

10. Hicks S, Candy DC, Phillips AD. 1996. Adhesion of enteroaggregativeEscherichia coli to pediatric intestinal mucosa in vitro. Infect. Immun.64:4751– 4760.

11. Jiang ZD, Greenberg D, Nataro JP, Steffen R, DuPont HL. 2002. Rateof occurrence and pathogenic effect of enteroaggregative Escherichia colivirulence factors in international travelers. J. Clin. Microbiol. 40:4185–4190. http://dx.doi.org/10.1128/JCM.40.11.4185-4190.2002.

12. Da Re S, Valle J, Charbonnel N, Beloin C, Latour-Lambert P, Faure P,Turlin E, Le Bouguénec C, Renauld-Mongénie G, Forestier C, GhigoJM. 2013. Identification of commensal Escherichia coli genes involved inbiofilm resistance to pathogen colonization. PLoS One 8:e61628. http://dx.doi.org/10.1371/journal.pone.0061628.

13. Roche JK, Cabel A, Sevilleja J, Nataro J, Guerrant RL. 2010.Enteroaggregative Escherichia coli (EAEC) impairs growth while malnu-trition worsens EAEC infection: a novel murine model of the infectionmalnutrition cycle. J. Infect. Dis. 202:506 –514. http://dx.doi.org/10.1086/654894.

14. Andrade JA, Freymüller E, Fagundes-Neto U. 2011. Adherence of entero-aggregative Escherichia coli to the ileal and colonic mucosa: an in vitrostudy utilizing the scanning electron microscopy. Arq. Gastroenterol.48:199 –204. http://dx.doi.org/10.1590/S0004-28032011000300009.

15. Sobieszczanska B, Duda AK, Turniak M, Duda-Madej A, Franiczek R,Kasprzykowska U. 2011. Characterization of genes associated with in-ternalization of enteroaggregative Escherichia coli. Microb. Pathog. 50:141–147. http://dx.doi.org/10.1016/j.micpath.2011.01.006.

16. Pereira AC, Britto-Filho JD, de Carvalho JJ, das Graças de Luna M,Rosa ACP. 2008. Enteroaggregative Escherichia coli (EAEC) strains enterand survive within cultured intestinal epithelial cells. Microb. Pathog.45:310 –314. http://dx.doi.org/10.1016/j.micpath.2008.07.001.

17. Thomazini CM, Samegima DA, Rodrigues MA, Victoria CR, Ro-drigues J. 2011. High prevalence of aggregative adherent Escherichia colistrains in the mucosa-associated microbiota of patients with inflamma-tory bowel diseases. Int. J. Med. Microbiol. 301:475– 479. http://dx.doi.org/10.1016/j.ijmm.2011.04.015.

18. Bhargava S, Johnson BB, Hwang J, Harris TA, George AS, Muir A,Dorff J, Okeke IN. 2009. Heat-resistant agglutinin 1 is an accessoryenteroaggregative Escherichia coli colonization factor. J. Bacteriol. 191:4934 – 4942. http://dx.doi.org/10.1128/JB.01831-08.

19. Czeczulin JR, Balepur S, Hicks S, Phillips A, Hall R, Kothary MH,Navarro-Garcia F, Nataro JP. 1997. Aggregative adherence fimbria II, asecond fimbrial antigen mediating aggregative adherence in enteroaggre-gative Escherichia coli. Infect. Immun. 65:4135– 4145.

20. Boisen N, Struve C, Scheutz F, Krogfelt KA, Nataro JP. 2008. Newadhesin of enteroaggregative Escherichia coli related to the Afa/Dr/AAFfamily. Infect. Immun. 76:3281–3292. http://dx.doi.org/10.1128/IAI.01646-07.

21. Nataro JP, Deng Y, Maneval DR, German AL, Martin WC, LevineMM. 1992. Aggregative adherence fimbriae I of enteroaggregative Esch-erichia coli mediate adherence to HEp-2 cells and hemagglutination ofhuman erythrocytes. Infect. Immun. 60:2297–2304.

22. Bernier C, Gounon P, Le Bouguénec C. 2002. Identification of anaggregative adhesion fimbria (AAF) type III-encoding operon in entero-aggregative Escherichia coli as a sensitive probe for detecting the AAF-encoding operon family. Infect. Immun. 70:4302– 4311. http://dx.doi.org/10.1128/IAI.70.8.4302-4311.2002.

23. Sheikh J, Czeczulin JR, Harrington S, Hicks S, Henderson IR, LeBouguénec C, Gounon P, Phillips A, Nataro JP. 2002. A novel dispersinprotein in enteroaggregative Escherichia coli. J. Clin. Invest. 110:1329 –1337. http://dx.doi.org/10.1172/JCI16172.

24. Farfan MJ, Inman KG, Nataro JP. 2008. The major pilin subunit of theAAF/II fimbriae from enteroaggregative Escherichia coli mediates bind-ing to extracellular matrix proteins. Infect. Immun. 76:4378 – 4384. http://dx.doi.org/10.1128/IAI.00439-08.

25. Izquierdo M, Navarro-Garcia F, Nava-Acosta R, Nataro JP, Ruiz-Perez

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 625

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 13: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

F, Farfan MJ. 2014. Identification of cell surface-exposed proteins in-volved in the fimbria-mediated adherence of enteroaggregative Esche-richia coli to intestinal cells. Infect. Immun. 82:1719 –1724. http://dx.doi.org/10.1128/IAI.01651-13.

26. Yamamoto T, Endo S, Yokota T, Echeverria P. 1991. Characteristics ofadherence of enteroaggregative Escherichia coli to human and animalmucosa. Infect. Immun. 59:3722–3739.

27. Morin N, Santiago AE, Ernst R K, Guillot S J, Nataro JP. 2013. Charac-terization of the AggR regulon in enteroaggregative Escherichia coli. Infect.Immun. 81:122–132. http://dx.doi.org/10.1128/IAI.00676-12.

28. Dudley EG, Thomson NR, Parkhill J, Morin NP, Nataro JP. 2006.Proteomic and microarray characterization of the AggR regulon identi-fies a pheU pathogenicity island in enteroaggregative Escherichia coli.Mol. Microbiol. 61:1267–1282. http://dx.doi.org/10.1111/j.1365-2958.2006.05281.x.

29. Huang DB, Mohamed JA, Nataro JP, DuPont HL, Jiang ZD,Okhuysen PC. 2007. Virulence characteristics and the molecular epide-miology of enteroaggregative Escherichia coli isolates from travellers todeveloping countries. J. Med. Microbiol. 56:1386 –1392. http://dx.doi.org/10.1099/jmm.0.47161-0.

30. Navarro-Garcia F, Gutierrez-Jimenez J, Garcia-Tovar C, Castro LA,Salazar-Gonzalez H, Cordova V. 2010. Pic, an autotransporter proteinsecreted by different pathogens in the Enterobacteriaceae family, is apotent mucus secretagogue. Infect. Immun. 78:4101– 4109. http://dx.doi.org/10.1128/IAI.00523-10.

31. Tokuda K, Nishi J, Imuta N, Fujiyama R, Kamenosono A, Manago K,Kawano Y. 2010. Characterization of typical and atypical enteroaggre-gative Escherichia coli in Kagoshima, Japan: biofilm formation and acidresistance. Microbiol. Immunol. 54:320 –329. http://dx.doi.org/10.1111/j.1348-0421.2010.00210.x.

32. Pereira AL, Ferraz LR, Silva RS, Giugliano LG. 2007. EnteroaggregativeEscherichia coli virulence markers: positive association with distinct clin-ical characteristics and segregation into 3 enteropathogenic E. coli sero-groups. J. Infect. Dis. 195:366 –374. http://dx.doi.org/10.1086/510538.

33. Sheikh J, Hicks S, Dall’Agnol M, Phillips AD, Nataro JP. 2001. Rolesfor Fis and YafK in biofilm formation by enteroaggregative Escherichiacoli. Mol. Microbiol. 41:983–997. http://dx.doi.org/10.1046/j.1365-2958.2001.02512.x.

34. Boll EJ, Struve C, Boisen N, Olesen B, Stahlhut SG, Krogfelt KA. 2013.Role of enteroaggregative Escherichia coli virulence factors in uropatho-genesis. Infect. Immun. 81:1164 –1171. http://dx.doi.org/10.1128/IAI.01376-12.

35. Shamir ER, Warthan M, Brown SP, Nataro JP, Guerrant RL, HoffmanPS. 2010. Nitazoxanide inhibits biofilm production and hemagglutina-tion by enteroaggregative Escherichia coli strains by blocking assembly ofAafA fimbriae. Antimicrob. Agents Chemother. 54:1526 –1533. http://dx.doi.org/10.1128/AAC.01279-09.

36. Fujiyama R, Nishi J, Imuta N, Tokuda K, Manago K, Kawano Y. 2008.The shf gene of a Shigella flexneri homologue on the virulent plasmidpAA2 of enteroaggregative Escherichia coli 042 is required for firm bio-film formation. Curr. Microbiol. 56:474 – 480. http://dx.doi.org/10.1007/s00284-008-9115-y.

37. Mohamed JA, Huang DB, Jiang ZD, DuPont HL, Nataro JP, Belkind-Gerson J, Okhuysen PC. 2007. Association of putative enteroaggrega-tive Escherichia coli virulence genes and biofilm production in isolatesfrom travelers to developing countries. J. Clin. Microbiol. 45:121–126.http://dx.doi.org/10.1128/JCM.01128-06.

38. Wani SA, Hussain I, Rather MA, Kabli ZA, Nagamani K, Nishikawa Y,Qureshi SD, Khan I. 2012. Putative virulence genes and biofilm pro-duction among typical enteroaggregative Escherichia coli isolates fromdiarrhoeic children in Kashmir and Andhra Pradesh. Indian J. Microbiol.52:587–592. http://dx.doi.org/10.1007/s12088-012-0284-9.

39. Mendez-Arancibia E, Vargas M, Soto S, Ruiz J, Kahigwa E, Schellen-berg D, Urassa H, Gascón J, Vila J. 2008. Prevalence of differentvirulence factors and biofilm production in enteroaggregative Esche-richia coli isolates causing diarrhea in children in Ifakara (Tanzania). Am.J. Trop. Med. Hyg. 78:985–989. http://www.ajtmh.org/content/78/6/985.long.

40. Nataro JP, Mai V, Johnson J, Blackwelder WC, Heimer R, Tirrell S,Edberg SC, Braden CR, Glenn Morris J, Jr, Hirshon JM. 2006. Diar-rheagenic Escherichia coli infection in Baltimore, Maryland, and NewHaven, Connecticut. Clin. Infect. Dis. 43:402– 407. http://dx.doi.org/10.1086/505867.

41. Henderson IR, Navarro-Garcia F, Nataro JP. 1998. The great escape:structure and function of the autotransporter proteins. Trends Micro-biol. 6:370 –378. http://dx.doi.org/10.1016/S0966-842X(98)01318-3.

42. Boisen N, Scheutz F, Rasko DA, Redman JC, Persson S, Simon J,Kotloff KL, Levine MM, Sow S, Tamboura B, Toure A, Malle D,Panchalingam S, Krogfelt KA, Nataro JP. 2012. Genomic characteriza-tion of enteroaggregative Escherichia coli from children in Mali. J. Infect.Dis. 205:431– 444. http://dx.doi.org/10.1093/infdis/jir757.

43. Benjelloun-Touimi Z, Sansonetti PJ, Parsot C. 1995. SepA, the majorextracellular protein of Shigella flexneri: autonomous secretion and in-volvement in tissue invasion. Mol. Microbiol. 17:123–135. http://dx.doi.org/10.1111/j.1365-2958.1995.mmi_17010123.x.

44. Navarro-García F, Sears C, Eslava C, Cravioto A, Nataro JP. 1999.Cytoskeletal effects induced by pet, the serine protease enterotoxin ofenteroaggregative Escherichia coli. Infect. Immun. 67:2184 –2192.

45. Eslava C, Navarro-García F, Czeczulin JR, Henderson IR, Cravioto A,Nataro JP. 1998. Pet, an autotransporter enterotoxin from enteroaggre-gative Escherichia coli. Infect. Immun. 66:3155–3163.

46. Guyer DM, Radulovic S, Jones F, Mobley HLT. 2002. Sat, the secretedautotransporter toxin of uropathogenic Escherichia coli, is a vacuolatingcytotoxin for bladder and kidney epithelial cells. Infect. Immun. 70:4539 – 4546. http://dx.doi.org/10.1128/IAI.70.8.4539-4546.2002.

47. Henderson IR, Czeczulin J, Eslava C, Noriega F, Nataro JP. 1999.Characterization of pic, a secreted protease of Shigella flexneri and en-teroaggregative Escherichia coli. Infect. Immun. 67:5587–5596.

48. Savarino SJ, McVeigh A, Watson J, Cravioto A, Molina J, EcheverriaP, Bhan MK, Levine MM, Fasano A. 1996. Enteroaggregative Esche-richia coli heat-stable enterotoxin is not restricted to enteroaggregative E.coli. J. Infect. Dis. 173:1019 –1022. http://dx.doi.org/10.1093/infdis/173.4.1019.

49. Savarino S, Farsano A, Robertson D, Levine MM. 1991. Enteroaggre-gative Escherichia coli elaborate a heat-stable enterotoxin demonstrablein an in vitro rabbit intestinal model. J. Clin. Invest. 87:1450 –1455.

50. Ménard L-P, Dubreuil JD. 2002. Enteroaggregative Escherichia coliheat-stable enterotoxin 1 (EAST1): a new toxin with an old twist. Crit.Rev. Microbiol. 28:43– 60. http://dx.doi.org/10.1080/1040-840291046687.

51. Huang DB, Jiang Z-D, Dupont HL. 2003. Association of virulencefactor-positive and -negative enteroaggregative Escherichia coli and oc-currence of clinical illness in travelers from the United States to Mexico.Am. J. Trop. Med. Hyg. 69:506 –508. http://www.ajtmh.org/content/69/5/506.long.

52. Aslani MM, Alikhani MY, Zavari A, Yousefi R, Zamani AR. 2011.Characterization of enteroaggregative Escherichia coli (EAEC) clinicalisolates and their antibiotic resistance pattern. Int. J. Infect. Dis. 15:e136 – e139. http://dx.doi.org/10.1016/j.ijid.2010.10.002.

53. Navarro-García F, Eslava C, Villaseca JM, López-Revilla R, CzeczulinJR, Srinivas S, Nataro JP, Cravioto A. 1998. In vitro effects of a high-molecular-weight heat-labile enterotoxin from enteroaggregative Esche-richia coli. Infect. Immun. 66:3149 –3154.

54. Fasano A, Noriega FR, Maneval DR, Jr, Chanasongcram S, Russell R,Guandalini S, Levine MM. 1995. Shigella enterotoxin 1: an enterotoxinof Shigella flexneri 2a active in rabbit small intestine in vivo and in vitro.J. Clin. Invest. 95:2853–2861. http://dx.doi.org/10.1172/JCI117991.

55. Nataro JP, Hicks S, Phillips AD, Vial PA, Sears CL. 1996. T84 cells inculture as a model for enteroaggregative Escherichia coli pathogenesis.Infect. Immun. 64:4761– 4768.

56. Vial PA, Robins-Browne R, Lior H, Prado V, Kaper JB, Nataro JP,Maneval D, Elsayed A, Levine MM. 1988. Characterization of enteroad-herent-aggregative Escherichia coli, a putative agent of diarrheal disease.J. Infect. Dis. 158:70 –79.

57. Schubert S, Rakin A, Karch H, Carniel E, Heesemann J. 1998. Preva-lence of the “high-pathogenicity island” of Yersinia species among Esch-erichia coli strains that are pathogenic to humans. Infect. Immun. 66:480 – 485.

58. Herzer PJ, Inouye S, Inouye M, Whittam TS. 1990. Phylogeneticdistribution of branched RNA-linked multicopy single-stranded DNAamong natural isolates of Escherichia coli. J. Bacteriol. 172:6175– 6181.

59. Czeczulin JR, Whittam TS, Henderson IR, Navarro-Garcia F, NataroJP. 1999. Phylogenetic analysis of enteroaggregative and diffusely adher-ent Escherichia coli. Infect. Immun. 67:2692–2699.

60. Escobar-Páramo P, Clermont O, Blanc-Potard AB, Bui H, Le Bougué-nec C, Denamur E. 2004. A specific genetic background is required for

Hebbelstrup Jensen et al.

626 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 14: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

acquisition and expression of virulence factors in Escherichia coli. Mol.Biol. Evol. 21:1085–1094. http://dx.doi.org/10.1093/molbev/msh118.

61. Clermont O, Bonacorsi S, Bingen E. 2000. Rapid and simple determi-nation of the Escherichia coli phylogenetic group. Appl. Environ. Micro-biol. 66:4555– 4558. http://dx.doi.org/10.1128/AEM.66.10.4555-4558.2000.

62. Okeke IN, Wallace-Gadsden F, Simons HR, Matthews N, Labar AS,Hwang J, Wain J. 2010. Multi-locus sequence typing of enteroaggrega-tive Escherichia coli isolates from Nigerian children uncovers multiplelineages. PLoS One 5:e14093. http://dx.doi.org/10.1371/journal.pone.0014093.

63. Nataro JP, Deng Y, Cookson S, Cravioto A, Savarino SJ, Guers LD,Levine MM, Tacket CO. 1995. Heterogeneity of enteroaggregative Esch-erichia coli virulence demonstrated in volunteers. J. Infect. Dis. 171:465–468. http://dx.doi.org/10.1093/infdis/171.2.465.

64. Mathewson JJ, Johnson PC, DuPont HL, Satterwhite TK, Winsor DK.1986. Pathogenicity of enteroadherent Escherichia coli in adult volun-teers. J. Infect. Dis. 154:524 –527. http://dx.doi.org/10.1093/infdis/154.3.524.

65. Jenkins C, Chart H, Willshaw GA, Cheasty T, Tompkins DS. 2007.Association of putative pathogenicity genes with adherence characteris-tics and fimbrial genotypes in typical enteroaggregative Escherichia colifrom patients with and without diarrhoea in the United Kingdom. Eur. J.Clin. Microbiol. Infect. Dis. 26:901–906. http://dx.doi.org/10.1007/s10096-007-0388-z.

66. Vial PA, Mathewson JJ, DuPont HL, Guers L, Levine MM. 1990.Comparison of two assay methods for patterns of adherence to HEp-2cells of Escherichia coli from patients with diarrhea. J. Clin. Microbiol.28:882– 885.

67. Sarantuya J, Nishi J, Wakimoto N, Erdene S, Nataro JP, Sheikh J,Iwashita M, Manago K, Tokuda K, Yoshinaga M, Miyata K, KawanoY. 2004. Typical enteroaggregative Escherichia coli is the most prevalentpathotype among E. coli strains causing diarrhea in Mongolian children.J. Clin. Microbiol. 42:133–139. http://dx.doi.org/10.1128/JCM.42.1.133-139.2004.

68. Elias WP, Uber AP, Tomita SK, Trabulsi LR, Gomes TA. 2002.Combinations of putative virulence markers in typical and variant en-teroaggregative Escherichia coli strains from children with and withoutdiarrhoea. Epidemiol. Infect. 129:49 –55. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2869874/pdf/12211596.pdf.

69. Moon JY, Park JH, Kim YB. 2005. Molecular epidemiological charac-teristics of virulence factors on enteroaggregative E. coli. FEMS Micro-biol. Lett. 253:215–220. http://dx.doi.org/10.1016/j.femsle.2005.09.038.

70. Cennimo DJ, Koo H, Mohamed JA, Huang DB, Chiang T. 2007.Enteroaggregative Escherichia coli: a review of trends, diagnosis, andtreatment. Infect. Med. 2007:100 –110. http://antimicrobe.org/h04c.files/history/E.%20Coli-Infect%20in%20Med%20-Cennimo%20et%20al%20article.pdf.

71. Nishi J, Sheikh J, Mizuguchi K, Luisi B, Burland V, Boutin A, Rose DJ,Blattner FR, Nataro JP. 2003. The export of coat protein from entero-aggregative Escherichia coli by a specific ATP-binding cassette transportersystem. J. Biol. Chem. 278:45680 – 45689. http://dx.doi.org/10.1074/jbc.M306413200.

72. Olesen B, Neimann J, Böttiger B, Ethelberg S, Schiellerup P, Jensen C,Helms M, Scheutz F, Olsen KE, Krogfelt K, Petersen E, Mølbak K,Gerner-Smidt P. 2005. Etiology of diarrhea in young children in Den-mark: a case-control study. J. Clin. Microbiol. 43:3636 –3641. http://dx.doi.org/10.1128/JCM.43.8.3636-3641.2005.

73. Jenkins C, Tembo M, Chart H, Cheasty T, Willshaw GA, PhillipsAD, Tompkins D, Smith H. 2006. Detection of enteroaggregativeEscherichia coli in faecal samples from patients in the community withdiarrhoea. J. Med. Microbiol. 55:1493–1497. http://dx.doi.org/10.1099/jmm.0.46683-0.

74. Regua-Mangia AH, Gomes TA, Vieira MA, Irino K, Teixeira LM. 2009.Molecular typing and virulence of enteroaggregative Escherichia colistrains isolated from children with and without diarrhoea in Rio deJaneiro city, Brazil. J. Med. Microbiol. 58:414 – 422. http://dx.doi.org/10.1099/jmm.0.006502-0.

75. Wilson A, Evans J, Chart H, Cheasty T, Wheeler JG, Tompkins D,Smith HR. 2001. Characterisation of strains of enteroaggregative Esche-richia coli isolated during the infectious intestinal disease study in Eng-land. Eur. J. Epidemiol. 17:1125–1130. http://dx.doi.org/10.1023/A:1021224915322.

76. Hien BT, Scheutz F, Cam PD, Serichantalergs O, Huong TT, Thu TM,Dalsgaard A. 2008. Diarrheagenic Escherichia coli and Shigella strainsisolated from children in a hospital case-control study in Hanoi, Viet-nam. J. Clin. Microbiol. 46:996 –1004. http://dx.doi.org/10.1128/JCM.01219-07.

77. Bellini EM, Elias WP, Gomes TA, Tanaka TL, Taddei CR, Huerta R,Navarro-Garcia F, Martinez MB. 2005. Antibody response against plas-mid-encoded toxin (Pet) and the protein involved in intestinal coloni-zation (Pic) in children with diarrhea produced by enteroaggregativeEscherichia coli. FEMS Immunol. Med. Microbiol. 43:259 –264. http://dx.doi.org/10.1016/j.femsim.2004.08.008.

78. Scavia G, Staffolani M, Fisichella S, Striano G, Colletta S, Ferri G, EscherM, Minelli F, Caprioli A. 2008. Enteroaggregative Escherichia coli associ-ated with a foodborne outbreak of gastroenteritis. J. Med. Microbiol.57:1141–1146. http://dx.doi.org/10.1099/jmm.0.2008/001362-0.

79. Harada T, Hiroi M, Kawamori F, Furusawa A, Ohata K, Sugiyama K,Masuda T. 2007. A food poisoning diarrhea outbreak caused by entero-aggregative Escherichia coli serogroup O126:H27 in Shizuoka, Japan. Jpn.J. Infect. Dis. 60:154 –155. http://www0.nih.go.jp/JJID/60/154.pdf.

80. Cobeljic M, Miljkovic-Selimovic B, Paunovic-Todosijevic D,Velickovic Z, Lepsanovic Z, Zec N, Savic D, Ilic R, Konstantinovic S,Jovanovic B, Kostic V. 1996. Enteroaggregative Escherichia coli associ-ated with an outbreak of diarrhoea in a neonatal nursery ward. Epide-miol. Infect. 117:11–16. http://dx.doi.org/10.1017/S0950268800001072.

81. Olesen B, Scheutz F, Andersen RL, Menard M, Boisen N, Johnston B,Hansen DS, Krogfelt KA, Nataro JP, Johnson JR. 2012. Enteroaggre-gative Escherichia coli O78:H10, the cause of an outbreak of urinary tractinfection. J. Clin. Microbiol. 50:3703–3711. http://dx.doi.org/10.1128/JCM.01909-12.

82. Spencer J, Smith HR, Chart H. 1999. Characterization of enteroaggre-gative Escherichia coli isolated from outbreaks of diarrhoeal disease inEngland. Epidemiol. Infect. 123:413– 421. http://dx.doi.org/10.1017/S0950268899002976.

83. Baudry B, Savarino SJ, Vial P, Kaper JB, Levine MM. 1990. A sensitiveand specific DNA probe to identify enteroaggregative Escherichia coli, arecently discovered diarrheal pathogen. J. Infect. Dis. 161:1249 –1251.http://dx.doi.org/10.1093/infdis/161.6.1249.

84. Kang G, Mathan M, Mathan V. 1995. Evaluation of a simplified HEp-2cell adherence assay for Escherichia coli isolated from south Indian chil-dren with acute diarrhea and controls. J. Clin. Microbiol. 33:2204 –2205.

85. Huang DB, Brown EL, DuPont HL, Cerf J, Carlin L, Flores J, Belkind-Gerson J, Nataro JP, Okhuysen PC. 2008. Seroprevalence of the entero-aggregative Escherichia coli virulence factor dispersin among USA trav-ellers to Cuernavaca, Mexico: a pilot study. J. Med. Microbiol. 57:476 –479. http://dx.doi.org/10.1099/jmm.0.47495-0.

86. Okhuysen PC, Jiang ZD, Carlin L, Forbes C, DuPont HL. 2004.Post-diarrhea chronic intestinal symptoms and irritable bowel syndromein North American travelers to Mexico. Am. J. Gastroenterol. 99:1774 –1778. http://dx.doi.org/10.1111/j.1572-0241.2004.30435.x.

87. Greenberg DE, Jiang ZD, Steffen R, Verenker MP, DuPont HL. 2002.Markers of inflammation in bacterial diarrhea among travelers, with afocus on enteroaggregative Escherichia coli pathogenicity. J. Infect. Dis.185:944 –949. http://dx.doi.org/10.1086/339617.

88. Shazberg G, Wolk M, Schmidt H, Sechter I, Gottesman G, Miron D.2003. Enteroaggregative Escherichia coli serotype O126:H27, Israel.Emerg. Infect. Dis. 9:1170 –1173. http://dx.doi.org/10.3201/eid0909.020695.

89. Abe CM, Knutton S, Pedroso MZ, Freymüller E, Gomes TA. 2001. Anenteroaggregative Escherichia coli strain of serotype O111:H12 damagesand invades cultured T84 cells and human colonic mucosa. FEMS Mi-crobiol. Lett. 203:199 –205. http://dx.doi.org/10.1111/j.1574-6968.2001.tb10841.x.

90. Itoh Y, Nagano I, Kunishima M, Ezaki T. 1997. Laboratory investiga-tion of enteroaggregative Escherichia coli O untypeable:H10 associatedwith a massive outbreak of gastrointestinal illness. J. Clin. Microbiol.35:2546 –2550.

91. Huang DB, DuPont HL. 2004. Enteroaggregative Escherichia coli: anemerging pathogen in children. Semin. Pediatr. Infect. Dis. 15:266 –271.http://dx.doi.org/10.1053/j.spid.2004.07.008.

92. Yatsuyanagi J, Saito S, Sato H, Miyajima Y, Amano K, Enomoto K.2002. Characterization of enteropathogenic and enteroaggregative Esch-erichia coli isolated from diarrheal outbreaks. J. Clin. Microbiol. 40:294 –297. http://dx.doi.org/10.1128/JCM.40.1.294-297.2002.

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 627

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 15: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

93. Steiner TS, Lima AA, Nataro JP, Guerrant RL. 1998. EnteroaggregativeEscherichia coli produce intestinal inflammation and growth impairmentand cause interleukin-8 release from intestinal epithelial cells. J. Infect.Dis. 177:88 –96. http://dx.doi.org/10.1086/513809.

94. Samie A, Obi CL, Dillingham R, Pinkerton RC, Guerrant RL. 2007.Enteroaggregative Escherichia coli in Venda, South Africa: distribution ofvirulence-related genes by multiplex polymerase chain reaction in stoolsamples of human immunodeficiency virus (HIV)-positive and HIV-negative individuals and primary school children. Am. J. Trop. Med.Hyg. 77:142–150. http://www.ajtmh.org/content/77/1/142.long.

95. Denno DM, Shaikh N, Stapp JR, Qin X, Hutter CM, Hoffman V,Mooney JC, Wood KM, Stevens HJ, Jones R, Tarr PI, Klein EJ. 2012.Diarrhea etiology in a pediatric emergency department: a case controlstudy. Clin. Infect. Dis. 55:897–904. http://dx.doi.org/10.1093/cid/cis553.

96. Scheutz F, Nielsen EM, Frimodt-Møller J, Boisen N, Morabito S,Tozzoli R, Nataro JP, Caprioli A. 2011. Characteristics of the entero-aggregative Shiga toxin/verotoxin-producing Escherichia coli O104:H4strain causing the outbreak of haemolytic uraemic syndrome in Ger-many, May to June 2011. Euro Surveill. 16(24):pii�19889. http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId�19889.

97. Beutin L, Martin A. 2012. Outbreak of Shiga toxin-producing Esche-richia coli (STEC) O104:H4 infection in Germany causes a paradigm shiftwith regard to human pathogenicity of STEC strains. J. Food Prot. 75:408 – 418. http://dx.doi.org/10.4315/0362-028X.JFP-11-452.

98. Rasko DA, Webster DR, Sahl JW, Bashir A, Boisen N, Scheutz F,Paxinos EE, Sebra R, Chin CS, Iliopoulos D, Klammer A, Peluso P, LeeL, Kislyuk AO, Bullard J, Kasarskis A, Wang S, Eid J, Rank D, RedmanJC, Steyert SR, Frimodt-Møller J, Struve C, Petersen AM, Krogfelt KA,Nataro JP, Schadt EE, Waldor MK. 2011. Origins of the E. coli straincausing an outbreak of hemolytic-uremic syndrome in Germany. N.Engl. J. Med. 365:709 –717. http://dx.doi.org/10.1056/NEJMoa1106920.

99. Bielaszewska M, Mellmann A, Zhang W, Köck R, Fruth A, Bauwens A,Peters G, Karch H. 2011. Characterisation of the Escherichia coli strainassociated with an outbreak of haemolytic uraemic syndrome in Ger-many, 2011: a microbiological study. Lancet Infect. Dis. 11:671– 676.http://dx.doi.org/10.1016/S1473-3099(11)70165-7.

100. Abe CM, Salvador FA, Falsetti IN, Vieira MA, Blanco J, Blanco JE,Blanco M, Machado AM, Elias WP, Hernandes RT, Gomes TA. 2008.Uropathogenic Escherichia coli (UPEC) strains may carry virulence prop-erties of diarrhoeagenic E. coli. FEMS Immunol. Med. Microbiol. 52:397– 406. http://dx.doi.org/10.1111/j.1574-695X.2008.00388.x.

101. Guyer DM, Henderson IR, Nataro JP, Mobley HL. 2000. Identificationof sat, an autotransporter toxin produced by uropathogenic Escherichiacoli. Mol. Microbiol. 38:53– 66. http://dx.doi.org/10.1046/j.1365-2958.2000.02110.x.

102. Herzog K, Engeler Dusel J, Hugentobler M, Beutin L, Sägesser G,Stephan R, Hächler H, Nüesch-Inderbinen M. 2014. Diarrheagenicenteroaggregative Escherichia coli causing urinary tract infection andbacteremia leading to sepsis. Infection 42:441– 444. http://dx.doi.org/10.1007/s15010-013-0569-x.

103. Khan K, Konar M, Goyal A, Ghosh S. 2010. Enteroaggregative Esche-richia coli infection induces IL-8 production via activation of mitogen-activated protein kinases and the transcription factors NF-kappaB andAP-1 in INT-407 cells. Mol. Cell. Biochem. 337:17–24. http://dx.doi.org/10.1007/s11010-009-0282-3.

104. Goyal A, Konar M, Setia A, Narang A, Ghosh S. 2010. Galactosespecific adhesin of enteroaggregative E. coli induces IL-8 secretion viaactivation of MAPK and STAT-3 in INT-407 cells. Biochim. Biophys.Acta 1800:574 –579. http://dx.doi.org/10.1016/j.bbagen.2010.03.009.

105. Opintan JA, Newman MJ, Ayeh-Kumi PF, Affrim R, Gepi-Attee R,Sevilleja JE, Roche JK, Nataro JP, Warren CA, Guerrant RL. 2010.Pediatric diarrhea in southern Ghana: etiology and association with in-testinal inflammation and malnutrition. Am. J. Trop. Med. Hyg. 83:936 –943. http://dx.doi.org/10.4269/ajtmh.2010.09-0792.

106. Steiner TS, Nataro JP, Poteet-Smith CE, Smith JA, Guerrant RL. 2000.Enteroaggregative Escherichia coli expresses a novel flagellin that causesIL-8 release from intestinal epithelial cells. J. Clin. Invest. 105:1769 –1777. http://dx.doi.org/10.1172/JCI8892.

107. Garcia PG, Silva VL, Diniz CG. 2011. Occurrence and antimicrobialdrug susceptibility patterns of commensal and diarrheagenic Escherichiacoli in fecal microbiota from children with and without acute diarrhea. J.Microbiol. 49:46 –52. http://dx.doi.org/10.1007/s12275-011-0172-8.

108. Amaya E, Reyes D, Vilchez S, Paniagua M, Mollby R, Nord CE,Weintraub A. 2011. Antibiotic resistance patterns of intestinal Esche-richia coli isolates from Nicaraguan children. J. Med. Microbiol. 60:216 –222. http://dx.doi.org/10.1099/jmm.0.020842-0.

109. Nüesch-Inderbinen MT, Hofer E, Hächler H, Beutin L, Stephan R.2013. Characteristics of enteroaggregative Escherichia coli isolated fromhealthy carriers and from patients with diarrhoea. J. Med. Microbiol.62(Part 12):1828 –1834. http://dx.doi.org/10.1099/jmm.0.065177-0.

110. Chaudhuri RR, Sebaihia M, Hobman JL, Webber MA, Leyton DL,Goldberg MD, Cunningham AF, Scott-Tucker A, Ferguson PR,Thomas CM, Frankel G, Tang CM, Dudley EG, Roberts IS, Rasko DA,Pallen MJ, Parkhill J, Nataro JP, Thomson NR, Henderson IR. 2010.Complete genome sequence and comparative metabolic profiling of theprototypical enteroaggregative Escherichia coli strain 042. PLoS One5:e8801. http://dx.doi.org/10.1371/journal.pone.0008801.

111. Mohamed JA, DuPont HL, Flores J, Palur H, Nair P, Jiang ZD, GuoD, Belkind-Gerson J, Okhuysen PC. 2011. Single nucleotide polymor-phisms in the promoter of the gene encoding the lipopolysaccharidereceptor CD14 are associated with bacterial diarrhea in US and Canadiantravelers to Mexico. Clin. Infect. Dis. 52:1332–1341. http://dx.doi.org/10.1093/cid/cir228.

112. Monteiro BT, Campos LC, Sircili MP, Franzolin MR, Bevilacqua LF,Nataro JP, Elias WP. 2009. The dispersin-encoding gene (aap) is notrestricted to enteroaggregative Escherichia coli. Diagn. Microbiol. Infect.Dis. 65:81– 84. http://dx.doi.org/10.1016/j.diagmicrobio.2009.05.011.

113. Sutjita M, Bouckenooghe AR, Adachi JA, Jiang ZD, Mathewson JJ,Ericsson CD, DuPont HL. 2000. Intestinal secretory immunoglobulin Aresponse to enteroaggregative Escherichia coli in travelers with diarrhea.Clin. Diagn. Lab. Immunol. 7:501–503. http://dx.doi.org/10.1128/CDLI.7.3.501-503.2000.

114. Gomez HF, Mathewson JJ, Johnson PC, DuPont HL. 1995. Intestinalimmune response of volunteers ingesting a strain of enteroadherent(HEp-2 cell-adherent) Escherichia coli. Clin. Diagn. Lab. Immunol. 2:10 –13.

115. Oundo JO, Kariuki SM, Boga HI, Muli FW, Iijima Y. 2008. Highincidence of enteroaggregative Escherichia coli among food handlers inthree areas of Kenya: a possible transmission route of travelers’ diarrhea.J. Travel Med. 15:31–38. http://dx.doi.org/10.1111/j.1708-8305.2007.00174.x.

116. Guiral E, Mendez-Arancibia E, Soto SM, Salvador P, Fabrega A,Gascon J, Vila J. 2011. CTX-M-15-producing enteroaggregative Esche-richia coli as cause of travelers’ diarrhea. Emerg. Infect. Dis. 17:1950 –1953. http://dx.doi.org/10.3201/eid1710.110022.

117. Vila J, Vargas M, Ruiz J, Espasa M, Pujol M, Corachán M, Jiménez deAnta MT, Gascón J. 2001. Susceptibility patterns of enteroaggregativeEscherichia coli associated with traveller’s diarrhoea: emergence ofquinolone resistance. J. Med. Microbiol. 50:996 –1000. http://jmm.sgmjournals.org/content/50/11/996.long.

118. Raju B, Ballal M. 2009. Multidrug resistant enteroaggregative Esche-richia coli diarrhoea in rural southern Indian population. Scand. J. Infect.Dis. 41:105–108. http://dx.doi.org/10.1080/00365540802641856.

119. Huppertz HI, Rutkowski S, Aleksic S, Karch H. 1997. Acute andchronic diarrhoea and abdominal colic associated with enteroaggregativeEscherichia coli in young children living in western Europe. Lancet 349:1660 –1662. http://dx.doi.org/10.1016/S0140-6736(96)12485-5.

120. Vasudevan P, Annamalai T, Sartori L, Hoagland T, VenkitanarayananK. 2003. Behavior of enteroaggregative Escherichia coli in bottled springand mineral water. J. Food Prot. 66:497–500.

121. Sidhu JPS, Ahmed W, Hodgers L, Toze S. 2013. Occurrence of viru-lence genes associated with diarrheagenic pathotypes in Escherichia coliisolates from surface water. Appl. Environ. Microbiol. 79:328 –335. http://dx.doi.org/10.1128/AEM.02888-12.

122. Akter S, Islam M, Afreen KS, Azmuda N, Khan SI, Birkeland NK.2013. Prevalence and distribution of different diarrhoeagenic Escherichiacoli virulotypes in major water bodies in Bangladesh. Epidemiol. Infect.141:2516 –2525. http://dx.doi.org/10.1017/S0950268813000320.

123. Kagambèga A, Martikainen O, Lienemann T, Siitonen A, Traoré AS,Barro N, Haukka K. 2012. Diarrheagenic Escherichia coli detected by16-plex PCR in raw meat and beef intestines sold at local markets inOuagadougou, Burkina Faso. Int. J. Food Microbiol. 153:154 –158. http://dx.doi.org/10.1016/j.ijfoodmicro.2011.10.032.

124. Morais TB, Gomes TA, Sigulem DM. 1997. Enteroaggregative Esche-

Hebbelstrup Jensen et al.

628 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 16: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

richia coli in infant feeding bottles. Lancet 349:1448 –1449. http://dx.doi.org/10.1016/S0140-6736(05)63724-5.

125. Cassar CA, Ottaway M, Paiba GA, Futter R, Newbould S, WoodwardMJ. 2004. Absence of enteroaggregative Escherichia coli in farmed ani-mals in Great Britain. Vet. Rec. 154:237–239. http://dx.doi.org/10.1136/vr.154.8.237.

126. Puño-Sarmiento J, Medeiros L, Chiconi C, Martins F, Pelayo J, RochaS, Blanco J, Blanco M, Zanutto M, Kobayashi R, Nakazato G. 2013.Detection of diarrheagenic Escherichia coli strains isolated from dogs andcats in Brazil. Vet. Microbiol. 166:676 – 680. http://dx.doi.org/10.1016/j.vetmic.2013.07.007.

127. Bibbal D, Kérourédan M, Loukiadis E, Scheutz F, Oswald E, BrugèreH. 2014. Slaughterhouse effluent discharges into rivers not responsiblefor environmental occurrence of enteroaggregative Escherichia coli. Vet.Microbiol. 168:451– 454. http://dx.doi.org/10.1016/j.vetmic.2013.11.042.

128. Johnson JJR, Owens K, Gajewski A, Clabots C. 2008. Escherichia colicolonization patterns among human household members and pets, withattention to acute urinary tract infection. J. Infect. Dis. 197:218 –224http://dx.doi.org/10.1086/524844.

129. Lima AA, Moore SR, Barboza MS, Jr, Soares AM, Schleupner MA,Newman RD, Sears CL, Nataro JP, Fedorko DP, Wuhib T, SchorlingJB, Guerrant RL. 2000. Persistent diarrhea signals a critical period ofincreased diarrhea burdens and nutritional shortfalls: a prospective co-hort study among children in northeastern Brazil. J. Infect. Dis. 181:1643–1645. http://dx.doi.org/10.1086/315423.

130. Dutta S, Pal S, Chakrabarti S, Dutta P, Manna B. 1999. Use of PCR toidentify enteroaggregative Escherichia coli as an important cause of acutediarrhoea among children living in Calcutta, India. J. Med. Microbiol.48:1011–1016. http://dx.doi.org/10.1099/00222615-48-11-1011.

131. Albert MJ, Faruque AS, Faruque SM, Sack RB, Mahalanabis D. 1999.Case-control study of enteropathogens associated with childhood diar-rhea in Dhaka, Bangladesh. J. Clin. Microbiol. 37:3458 –3464.

132. Albert MJ, Faruque SM, Faruque AS, Neogi PK, Ansaruzzaman M,Bhuiyan NA, Alam K, Akbar MS. 1995. Controlled study of Escherichiacoli diarrheal infections in Bangladeshi children. J. Clin. Microbiol. 33:973–977.

133. Rajendran P, Ajjampur SS, Chidambaram D, Chandrabose G, Thanga-raj B, Sarkar R, Samuel P, Rajan DP, Kang G. 2010. Pathotypes ofdiarrheagenic Escherichia coli in children attending a tertiary care hospi-tal in South India. Diagn. Microbiol. Infect. Dis. 68:117–122. http://dx.doi.org/10.1016/j.diagmicrobio.2010.06.003.

134. Durrer P, Zbinden R, Fleisch F, Altwegg M, Ledergerber B, Karch H,Weber R. 2000. Intestinal infection due to enteroaggregative Escherichiacoli among human immunodeficiency virus-infected persons. J. Infect.Dis. 182:1540 –1544. http://dx.doi.org/10.1086/315885.

135. Gassama-Sow A, Sow PS, Guèye M, Guèye-N=diaye A, Perret JL,M’boup S, Aïdara-Kane A. 2004. Characterization of pathogenic Esch-erichia coli in human immunodeficiency virus-related diarrhea in Sene-gal. J. Infect. Dis. 189:75–78. http://dx.doi.org/10.1086/380489.

136. Schmidt H, Knop C, Franke S, Aleksic S, Heesemann J, Karch H. 1995.Development of PCR for screening of enteroaggregative Escherichia coli.J. Clin. Microbiol. 33:701–705.

137. Medina AM, Rivera FP, Romero LM, Kolevic LA, Castillo ME, VerneE, Hernandez R, Mayor YE, Barletta F, Mercado E, Ochoa TJ. 2010.Diarrheagenic Escherichia coli in human immunodeficiency virus (HIV)pediatric patients in Lima, Peru. Am. J. Trop. Med. Hyg. 83:158 –163.http://dx.doi.org/10.4269/ajtmh.2010.09-0596.

138. Paschke C, Apelt N, Fleischmann E, Perona P, Walentiny C, LöscherT, Herbinger KH. 2011. Controlled study on enteropathogens in trav-ellers returning from the tropics with and without diarrhoea. Clin. Mi-crobiol. Infect. 17:1194 –1200. http://dx.doi.org/10.1111/j.1469-0691.2010.03414.x.

139. Adachi JA, Jiang ZD, Mathewson JJ, Verenkar MP, Thompson S,Martinez-Sandoval F, Steffen R, Ericsson CD, DuPont HL. 2001.Enteroaggregative Escherichia coli as a major etiologic agent in traveler’sdiarrhea in 3 regions of the world. Clin. Infect. Dis. 32:1706 –1709. http://dx.doi.org/10.1086/320756.

140. Lääveri T, Pakkanen SH, Antikainen J, Riutta J, Mero S, Kirveskari J,Kantele A. 2014. High number of diarrhoeal coinfections in travellers toBenin, West Africa. BMC Infect. Dis. 14:81. http://dx.doi.org/10.1186/1471-2334-14-81.

141. Ahn JY, Chung JW, Chang KJ, You MH, Chai JS, Kang YA, Kim SH,

Jeoung H, Cheon D, Jeoung A, Choi ES. 2011. Clinical characteristicsand etiology of travelers’ diarrhea among Korean travelers visitingSouth-East Asia. J. Korean Med. Sci. 26:196 –200. http://dx.doi.org/10.3346/jkms.2011.26.2.196.

142. Vernacchio L, Vezina RM, Mitchell AA, Lesko SM, Plaut AG, AchesonDW. 2006. Diarrhea in American infants and young children in thecommunity setting. Pediatr. Infect. Dis. J. 25:2–7. http://dx.doi.org/10.1097/01.inf.0000195623.57945.87.

143. Cohen MB, Nataro JP, Bernstein DI, Hawkins J, Roberts N, Staat MA.2005. Prevalence of diarrheagenic Escherichia coli in acute childhood en-teritis: a prospective controlled study. J. Pediatr. 146:54 – 61. http://dx.doi.org/10.1016/j.jpeds.2004.08.059.

144. Chattaway MA, Harris R, Jenkins C, Tam C, Coia JE, Gray J, Iturriza-Gomara M, Wain J. 2013. Investigating the link between the presence ofenteroaggregative Escherichia coli and infectious intestinal disease in theUnited Kingdom, 1993 to 1996 and 2008 to 2009. Euro Surveill. 18(37):pii�20582. http://www.eurosurveillance.org/ViewArticle.aspx?ArticleId�20582.

145. Park HK, Jung YJ, Chae HC, Shin YJ, Woo SY, Park HS, Lee SJ. 2009.Comparison of Escherichia coli uropathogenic genes (kps, usp and ireA)and enteroaggregative genes (aggR and aap) via multiplex polymerasechain reaction from suprapubic urine specimens of young children withfever. Scand. J. Urol. Nephrol. 43:51–57. http://dx.doi.org/10.1080/00365590802299338.

146. Buchholz U, Bernard H, Werber D, Böhmer MM, Remschmidt C,Wilking H, Deleré Y, an der Heiden M, Adlhoch C, Dreesman J, EhlersJ, Ethelberg S, Faber M, Frank C, Fricke G, Greiner M, Höhle M,Ivarsson S, Jark U, Kirchner M, Koch J, Krause G, Luber P, Rosner B,Stark K, Kühne M. 2011. German outbreak of Escherichia coli O104:H4associated with sprouts. N. Engl. J. Med. 365:1763–1770. http://dx.doi.org/10.1056/NEJMoa1106482.

147. Morabito S, Karch H, Mariani-Kurkdjian P, Schmidt H, Minelli F,Bingen E, Caprioli A. 1998. Enteroaggregative, Shiga toxin-producingEscherichia coli O111:H2 associated with an outbreak of hemolytic-uremic syndrome. J. Clin. Microbiol. 36:840 – 842.

148. Iyoda S, Tamura K, Itoh K, Izumiya H, Ueno N, Nagata K, Togo M,Terajima J, Watanabe H. 2000. Inducible stx2 phages are lysogenized inthe enteroaggregative and other phenotypic Escherichia coli O86:HNMisolated from patients. FEMS Microbiol. Lett. 191:7–10. http://dx.doi.org/10.1111/j.1574-6968.2000.tb09311.x.

149. Dallman T, Smith GP, O’Brien B, Chattaway MA, Finlay D, Grant KA,Jenkins C. 2012. Characterization of a verocytotoxin-producing entero-aggregative Escherichia coli serogroup O111:H21 strain associated with ahousehold outbreak in Northern Ireland. J. Clin. Microbiol. 50:4116 –4119. http://dx.doi.org/10.1128/JCM.02047-12.

150. Boudailliez B, Berquin P, Mariani-Kurkdjian P, Ilef D, Cuvelier B,Capek I, Tribout B, Bingen E, Piussan C. 1997. Possible person-to-person transmission of Escherichia coli O111-associated hemolytic ure-mic syndrome. Pediatr. Nephrol. 11:36 –39. http://dx.doi.org/10.1007/s004670050229.

151. Pabst WL, Altwegg M, Kind C, Hardegger D, Nadal D, Mirjanic S.2003. Prevalence of enteroaggregative Escherichia coli among childrenwith and without diarrhea in Switzerland. J. Clin. Microbiol. 41:2289 –2293. http://dx.doi.org/10.1128/JCM.41.6.2289-2293.2003.

152. Pass MA, Odedra R, Batt RM. 2000. Multiplex PCRs for identificationof Escherichia coli virulence genes. J. Clin. Microbiol. 38:2001–2004. http://jcm.asm.org/content/38/5/2001.long.

153. Zamboni A, Fabbricotti SH, Fagundes-Neto U, Scaletsky ICA. 2004.Enteroaggregative Escherichia coli virulence factors are found to be as-sociated with infantile diarrhea in Brazil. J. Clin. Microbiol. 42:1058 –1063. http://dx.doi.org/10.1128/JCM.42.3.1058-1063.2004.

154. Cennimo D, Abbas A, Huang DB, Chiang T. 2009. The prevalence andvirulence characteristics of enteroaggregative Escherichia coli at an ur-gent-care clinic in the USA: a case-control study. J. Med. Microbiol.58:403– 407. http://dx.doi.org/10.1099/jmm.0.005793-0.

155. Jiang ZD, Okhuysen PC, Guo DC, He R, King TM, DuPont HL,Milewicz DM. 2003. Genetic susceptibility to enteroaggregative Esche-richia coli diarrhea: polymorphism in the interleukin-8 promotor region.J. Infect. Dis. 188:506 –511. http://dx.doi.org/10.1086/377102.

156. Taniuchi M, Walters CC, Gratz J, Maro A, Kumburu H, Serichanta-lergs O, Sethabutr O, Bodhidatta L, Kibiki G, Toney DM, Berkeley L,Nataro JP, Houpt ER. 2012. Development of a multiplex polymerasechain reaction assay for diarrheagenic Escherichia coli and Shigella spp.

Epidemiology of EAEC

July 2014 Volume 27 Number 3 cmr.asm.org 629

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from

Page 17: Epidemiology and Clinical Manifestations of ...fectious diarrhea is the so-called diarrheagenic Escherichia coli (DEC) group (1, 2). The following subgroups of DEC have been defined:enterotoxigenicE.coli(ETEC),Shigatoxin-producingE

and its evaluation on colonies, culture broths, and stool. Diagn. Micro-biol. Infect. Dis. 73:121–128. http://dx.doi.org/10.1016/j.diagmicrobio.2012.03.008.

157. Lima IFN, Boisen N, da Quetz Silva J, Havt A, de Carvalho EB, Soares AM,Lima NL, Mota RMS, Nataro JP, Guerrant RL, Lima AAM. 2013. Prevalenceof enteroaggregative Escherichia coli and its virulence-related genes in acase-control study among children from north-eastern Brazil. J. Med.Microbiol. 62:683– 693. http://dx.doi.org/10.1099/jmm.0.054262-0.

158. Meng CY, Smith BL, Bodhidatta L, Richard SA, Vansith K, Thy B, SrijanA, Serichantalergs O, Mason CJ. 2011. Etiology of diarrhea in young chil-

dren and patterns of antibiotic resistance in Cambodia. Pediatr. Infect. Dis. J.30:331–335. http://dx.doi.org/10.1097/INF.0b013e3181fb6f82.

159. Huppertz H, Busch D, Schmidt H, Aleksic S, Karch H. 1996. Diarrheain young children associated with Escherichia coli non-O 157 organismsthat produce Shiga-like toxin. J. Paediatr. 128:341–346. http://dx.doi.org/10.1016/S0022-3476(96)70278-0.

160. Tompkins DS, Hudson MJ, Smith HR, Eglin RP, Wheeler JG, BrettMM, Owen RJ, Brazier JS, Cumberland P, King V, Cook PE. 1999. Astudy of infectious intestinal disease in England: microbiological findingsin cases and controls. Commun. Dis. Public Health 2:108 –113.

Carsten Struve, M.Sc., Ph.D., is a Senior Scien-tist at the Department of Microbiology and In-fection Control, Statens Serum Institut, Den-mark. He did his M.Sc. studies at the Universityof Copenhagen and completed his Ph.D. in mo-lecular microbiology in 2002. His research fo-cuses on the identification and characterizationof virulence and resistance mechanisms in En-terobacteriaceae, including the developmentand use of experimental infection models.

Katharina E. P. Olsen, Ph.D., Pharm.D., is theHead of the National Reference Laboratory forEnteropathogenic Bacteria at the Departmentof Microbiology and Infection Control, StatensSerum Institut (SSI), Copenhagen, Denmark,and is responsible for the national surveillanceof Clostridium difficile. Formerly, she held aone-year position as a researcher at the WHOInternational Collaborating Centre for Refer-ence and Research on Escherichia and Klebsiella,SSI. She was educated at the School of Pharma-ceutical Sciences, Faculty of Health and Medical Sciences, Copenhagen Uni-versity, where she also earned her Ph.D. in medical molecular biology. Dr.Olsen is an officially appointed examiner in Microbiology at CopenhagenUniversity, Faculty of Health and Medical Sciences. She is a member of theDanish Society for Clinical Microbiology, the Danish National Committeeunder the Nordic Committee on Food Analysis, and the ESCMID StudyGroup for Clostridium difficile (ESGCD). Her research focuses on molecularepidemiology, diagnostics, and cytotoxicity of bacterial protein toxins.

Betina Hebbelstrup Jensen qualified in medi-cine at Aarhus University, Denmark, and Co-penhagen University, Denmark, in 2010. Part ofher medical internship was conducted at theDepartment of Gastroenterology, GlostrupHospital, Copenhagen, Denmark. Currently,she is performing her Ph.D. studies at StatensSerum Institut, Department of Microbiologyand Infection Control, on enteroaggregativeEscherichia coli, with a focus on the clinical im-plications associated with this microorganism.

Andreas Munk Petersen completed his M.D.and Ph.D. in Gastroenterology at the Universityof Copenhagen. He completed a residency ininternal medicine and gastroenterology at Her-lev University Hospital, Denmark, before con-tinuing his employment as a consultant at Hvi-dovre University Hospital, Denmark. Hisresearch focuses on the interfaces between mi-crobiology and gastroenterology.

Karen Angeliki Krogfelt, M.Sc. (Eng.), Ph.D.,is an adjunct professor at the Institute of Sys-tems Biology, Technical University of Den-mark, DTU, and Head of a research unit at theDepartment of Microbiology, Statens SerumInstitut. She is involved in teaching master’scourses and has supervised numerous master’sand Ph.D. students. Her main research activi-ties focus on the pathogenic mechanisms re-lated mainly to intestinal bacterial infectionsand on assessing the development of gut micro-biota and its relation to pathogenesis. Characterization of specific virulencefactors involved in pathogenesis is also used for the development of diagnos-tic methods for bacterial infections.

Hebbelstrup Jensen et al.

630 cmr.asm.org Clinical Microbiology Reviews

on May 20, 2021 by guest

http://cmr.asm

.org/D

ownloaded from