Next Article in Journal
Designing Statistical Models for Holstein Rearing Heifers’ Weight Estimation from Birth to 15 Months Old Using Body Measurements
Next Article in Special Issue
Diversity of Non-O157 Shiga Toxin-Producing Escherichia coli Isolated from Cattle from Central and Southern Chile
Previous Article in Journal
Relationship between Temperament and Stage of Lactation, Productivity and Milk Composition of Dairy Cows
Previous Article in Special Issue
Screening the Presence of Non-Typhoidal Salmonella in Different Animal Systems and the Assessment of Antimicrobial Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genomic Epidemiology of Shiga Toxin-Producing Escherichia coli Isolated from the Livestock-Food-Human Interface in South America

1
Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
2
Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
3
Facultad de Ciencias Agropecuarias y Ambientales, Universidad Pedro de Valdivia, Santiago 8370007, Chile
4
Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
5
Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA
6
Departamento de Patología, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
7
Departamento de Microbiología, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brazil
*
Author to whom correspondence should be addressed.
Animals 2021, 11(7), 1845; https://doi.org/10.3390/ani11071845
Submission received: 11 May 2021 / Revised: 15 June 2021 / Accepted: 16 June 2021 / Published: 22 June 2021
(This article belongs to the Special Issue The Impact of Emerging Hazards in Animal Production)

Abstract

:

Simple Summary

Shiga toxin-producing Escherichia coli (STEC) are zoonotic pathogens that cause food-borne diseases in humans, where cattle and derived products play a key role as reservoirs and vehicles. We analyzed the genomic data of STEC strains circulating at the livestock-food-human interface in South America, extracting clinically and epidemiologically relevant information (serotypes, virulome, resistance genes, sequence types, and phylogenomics). This study included 130 STEC genomes obtained from cattle (n = 51), beef (n = 48), and human (n = 31) samples. The successful expansion of O157:H7 (ST11) and non-O157 (ST16, ST21, ST223, ST443, ST677, ST679, ST2388) clones is highlighted, suggesting common activities, such as multilateral trade and travel. Circulating STEC strains analyzed exhibit high genomic diversity and harbor several genetic determinants associated with severe illness in humans, highlighting the need to establish official surveillance of this pathogen that should be focused on detecting molecular determinants of virulence and clonal relatedness, in the whole beef production chain.

Abstract

Shiga toxin-producing Escherichia coli (STEC) are zoonotic pathogens responsible for causing food-borne diseases in humans. While South America has the highest incidence of human STEC infections, information about the genomic characteristics of the circulating strains is scarce. The aim of this study was to analyze genomic data of STEC strains isolated in South America from cattle, beef, and humans; predicting the antibiotic resistome, serotypes, sequence types (STs), clonal complexes (CCs) and phylogenomic backgrounds. A total of 130 whole genome sequences of STEC strains were analyzed, where 39.2% were isolated from cattle, 36.9% from beef, and 23.8% from humans. The ST11 was the most predicted (20.8%) and included O-:H7 (10.8%) and O157:H7 (10%) serotypes. The successful expansion of non-O157 clones such as ST16/CC29-O111:H8 and ST21/CC29-O26:H11 is highlighted, suggesting multilateral trade and travel. Virulome analyses showed that the predominant stx subtype was stx2a (54.6%); most strains carried ehaA (96.2%), iha (91.5%) and lpfA (77.7%) genes. We present genomic data that can be used to support the surveillance of STEC strains circulating at the livestock-food-human interface in South America, in order to control the spread of critical clones “from farm to table”.

1. Introduction

Shiga toxin-producing Escherichia coli (STEC) is a zoonotic diarrheagenic pathotype of E. coli with the common characteristic of producing cytotoxins of the Shiga toxin family (Stx) [1]. Although their participation in different diarrheal processes in animals is recognized, their frequency is low; therefore, they are considered as reservoirs and disseminators of STEC. Thus, gut colonization favors transmission to other animals, the environment, and humans [2].
Cattle, which lack cellular receptors for Stx [3], are asymptomatic carriers of STEC strains and are recognized as the main source for human STEC infections. Most outbreaks are associated with the consumption of undercooked beef or unpasteurized dairy products [4]. In addition, some animals known as “super-shedders” can excrete more than 104 CFU/g feces [5], increasing the probability of infection in other animals and contamination of meat. Reported prevalence of STEC in cattle in the last 10 years in South America has ranged from 14% to 90% [6,7,8,9]. Given that the presence of this pathogen is frequent in cattle feces, their carcasses may become contaminated during slaughtering operations, leading to meat products containing STEC. Thus, one of the most common STEC vehicles associated with outbreaks is beef [10,11]. Therefore, preventing the introduction of this pathogen into the food chain represents a serious challenge for food safety and health official agencies [12]. In this context, in the USA serogroups O26, O45, O103, O111, O121, and O145 (also known as “the big six”), alongside O157 have been considered adulterants in raw, non-intact ground beef products since 2011 [13]. Nevertheless, the presence of STEC in soil, water, and animal manure are factors associated with the contamination of other foods such as vegetables [14,15].
In humans, STEC infections can cause severe gastroenteritis, hemorrhagic colitis (HC), and life-threatening hemolytic-uremic syndrome (HUS), mainly in children under five years of age [16]. Foodborne STEC infections cause more than one million cases of illness worldwide per year, with approximately 3890 HUS cases and 230 deaths [17,18]. In South America, STEC infections remain endemic and have contributed to the burden of acute diarrheal syndrome in this region, with a significant number of HUS cases mainly in Argentina, Chile, Uruguay, and Brazil [19,20,21]. This could be explained by the high beef consumption in those countries [22]; the cattle biomass in South America, which in 2018 was 357,222,668 animals, mainly concentrated in Brazil (59.8%), Argentina (15.1%), and Colombia (7.2%) [23]; and by the presence of hypervirulent clones circulating in the cattle-beef-human interface [24]. In this sense, a previous study in Argentina characterized a collection of STEC O157 strains isolated from cattle and humans; reporting that 98% of the strains harbored molecular determinants associated with severe illness in humans [25], a situation not reported anywhere before. Nevertheless, there are no other studies addressing the phylogenomics of STEC strains in South America; a situation that highlights the need to conduct robust analysis that can characterize and geographically track STEC strains circulating in the animal-food-human interface, as a strategy to promote animal and public health, under the concept of One Health.
Although epidemiological studies on STEC distribution have shown that O157:H7 serotype has been most frequently associated with outbreaks [26], “big-six” non-O157 STEC serogroups have also been associated with severe illness in humans worldwide [5]. However, most STEC strains isolated from cattle in South American countries do not belong to the aforementioned serogroups [8,27,28,29,30]. Given these contradictory reports, the isolation and serotype rates of STEC strains isolated from cattle are insufficient to explain the high HUS incidence in South American countries [19]. Moreover, a high genotypic diversity has been reported within serotypes [12], and horizontal gene transfer of the O-antigen can occur among different E. coli strains [31], making the prediction of the virulence of a strain based on its serotype inaccurate [32]. Therefore, additional analyses are required, such as virulome, resistome, and MLST patterns of the circulating strains.
The primary virulence factor of STEC is Stx, which is classified into two types, Stx1 and Stx2. Each type is further classified into several subtypes (stx1a, stx1c-e, stx2a-l) [33]. The typing and subtyping of Stx is necessary to predict the virulence of the strain, as Stx2 has been shown to be more virulent than Stx1, and is more frequently associated with HC and HUS, as well as the Stx2a, Stx2c, or Stx2d subtypes [5]. Additionally, STEC may harbor other virulence factors that contribute to the development of severe illness in humans, such as the eae gene, which is located in the locus of enterocyte effacement (LEE) [5]; or the saa gene [34] and the locus of adhesion and autoaggregation (LAA) [35] in the LEE-negative strains. More recently, convergence of virulence and antimicrobial resistance (AMR) among STEC strains has emerged as a worrying problem that threatens animal and public health globally. Indeed, STEC strains exhibiting a multidrug-resistant profile have been identified worldwide, including South America [36,37,38,39,40,41]. International trade of food and animals as well as international travelers can lead to the introduction of highly virulent and antimicrobial resistant strains [12,42,43,44], modifying the traditional epidemiology of the pathogen, and challenging biosecurity and surveillance protocols.
Given the high variety of serogroups, virulotypes and resistotypes, and the intrinsic genomic flexibility of E. coli, whole genome sequencing (WGS) is a specific and sensitive method to conduct epidemiological and phylogeographic studies. Several genomic studies have been carried out to determine the virulome, resistome, and the worldwide spread of STEC clones isolated from animals, the environment, food, and humans [32,45,46,47]. In South American countries these studies are scarce, and have been mainly focused on virulence determinants associated with HC and HUS [35,48].
Genomic surveillance of STEC strains is necessary to improve the knowledge about the dynamic of dissemination, and of the role of cattle as reservoirs and disseminators of STEC lineages associated with severe disease in humans. Thus, the aim of this study was to analyze genomic data of STEC strains isolated from cattle in Chile, and to perform a comparative genomic analysis of STEC strains circulating at the cattle-beef-human interface in South America, extracting clinically and epidemiologically relevant information, including virulome, antimicrobial resistance genes, sequence types, clonal complexes, and phylogenomics, in order to provide relevant epidemiological data to surveillance programs at national and international level, under a One Health approach.

2. Materials and Methods

2.1. Bacterial Isolates and Whole-Genome Sequencing (WGS)

Twenty-one STEC strains isolated from cattle were sequenced in this study, of which twenty were isolated in 2018 from cattle at abattoirs in the Región Metropolitana of Chile [8], and one was isolated in 2019 from cattle raised in a backyard production system in the same region [49]. Genomic DNA of these 21 STEC strains was extracted using the Wizard Genomic DNA purification kit (Promega, Madison, WI, USA), following the manufacturer’s instructions. Genomic DNA libraries were constructed using the QIAseq FX DNA library kit (Qiagen, Hilden, Germany). Sequencing was performed using the MiSeq Reagent kit v3 600 cycles on an Illumina MiSeq platform (Illumina, San Diego, CA, USA). All raw FASTQ files were uploaded and processed through the QAssembly (v3.61) automated assembly pipeline on the Enterobase platform (http://enterobase.warwick.ac.uk). De novo assemblies that passed quality control with the standard established by Enterobase for E. coli/Shigella were used [50]. Contigs shorter than 200 nt were removed and sequences were deposited at GenBank under BioProject numbers PRJNA656305 and PRJNA682583.

2.2. Publicly Available Genome Sequences

For comparative analysis, all E. coli strains isolated in any South American country from cattle, beef, and people, whose FASTQ files were available at SRA GenBank (https://www.ncbi.nlm.nih.gov/genbank/) were downloaded on 6 November 2020. These 600 whole genome sequences were mapped using BWA [51] on the sequences of every stx type/subtype. Thus, a total of 109 genomes of stx-positive E. coli strains were selected for comparative analysis with the 21 STEC genomes obtained in this study (Table S1).

2.3. Virulome and AMR Gene Analysis

The prediction of virulence genes was performed on VirulenceFinder 2.0 (https://cge.cbs.dtu.dk/services/VirulenceFinder/) and ABRicate (v.0.8.13) (https://github.com/tseemann/abricate/). The assessed virulence genes included aggR (aggregative adherence transcriptional regulator), cdtB (cytolethal distending toxin), eae (intimin), efa1(adhesion and invasion of epithelial cells), ehaA (adhesion to epithelial cells), ehxA (enterohemolysin), hlyA (α-hemolysin), hra (intimate attachment to microvilli), iha (adhesion and invasion of epithelial cells), lpfA (adhesion to fibronectin, laminin, and collagen), subA (subtilase cytotoxin), tia (adhesion and invasion of epithelial cells), and stx subtypes. In the case of saa and sab (both related to adhesion to epithelial cells) genes, as well as LAA and some of its associated genes, including ag43 (adhesion, autoaggregation, and biofilm formation), hes (adherence to epithelial cells), lesP (serine protease), and pagC-like (putative serum resistance), were screened through BLASTn (https://blast.ncbi.nlm.nih.gov/Blast.cgi), according to protocols described in a previous study [48]. ResFinder 4.1 (https://cge.cbs.dtu.dk/services/ResFinder/) was used to identify the AMR genes. These analyses were performed with a default setting of 90% of identity threshold and 60% minimum gene length overlap. The presence of these genes was confirmed when a coverage and identity >90% was achieved.

2.4. Epidemiological Typing and Phylogenomic Analysis

The multilocus sequence typing (MLST) scheme of Achtman was used to identify sequence types (STs) and clonal complexes (CCs) on the Enterobase platform (http://enterobase.warwick.ac.uk). We have additionally performed a systematic search of scientific literature containing MLST data of STEC strains identified in South American countries. From genome and literature data, we have constructed a map containing the distribution of STs of STEC strains circulating at the livestock-beef-environment-human interface in South America [21,39,52,53,54,55,56]. The SerotypeFinder 2.0 tool (https://cge.cbs.dtu.dk/services/SerotypeFinder/) was used to determine serotype [57]. A minimum spanning tree was constructed in Enterobase with the MSTree V2 algorithm and the wgMLST scheme (http://enterobase.warwick.ac.uk) for phylogenomic analysis of all STEC genomes. The phylogenetic tree was generated with interactive Tree of Life, iTOL v6 (https://itol.embl.de). CSI Phylogeny 1.4 (https://cge.cbs.dtu.dk/services/CSIPhylogeny) was used with default settings to generate an approximately maximum-likelihood phylogenetic tree with the 21 genome assemblies obtained from this study, using E. coli strain K-12 MG1655 (RefSeq accession number NC_000913.3) as reference genome. The percentage of reference genome covered by all isolates was 81.24%, corresponding to 3,771,065 positions found in all genomes. iTOL v6 (https://itol.embl.de) was used to root the tree at midpoint and annotate the tree with data from Enterobase and genomic data.

2.5. Statistical Analysis

A logistic multivariable regression model was performed to assess the association between stx type (dependent variable) and the geographic location and isolation source of the strains included in this study (independent variables), where Y (response) can have only two values (Y = 0 or Y = 1) [58]. Three models were developed, one for each stx type gene and one for the presence of both stx types at the same time. To ensure adequate convergence of the model and increase the statistical power of the results, epsilon was set at a high level (epsilon = 1 × 10−6). The fit of the models to the data was evaluated with the Hosmer–Lemeshow test [59]. The variable geographic location was modified to fit into the analysis by collapsing the isolates from Uruguay, Paraguay, and Ecuador into a new variable defined as other. All the statistical analyses were performed using R 4.0.2 (The R Foundation, Vienna, Austria) [60].

3. Results

3.1. Source and Origin of the Analyzed STEC Genomes

In total 130 STEC genomes were analyzed, of which 21 were obtained in this study. Genomes were representative of STEC isolates recovered from cattle (39.2%), beef (36.9%), and humans (23.8%). Analyzed STEC genomes were from Chile (n = 88), Argentina (n = 30), Ecuador (n = 7), Paraguay (n = 3), and Uruguay (n = 2). Table S2 contains the source, origin, and isolation date of each analyzed STEC strain.

3.2. Virulome and Antibiotic Resistome of STEC Strains

All strains met the WGS quality parameters of Enterobase, with a total average of filtered reads of 419,230.8 and an average coverage of 28.6 (Table S1). Among the 130 STEC genomes, 72.3% (n = 94) harbored the stx2 gene alone, 20.8% (n = 27) were stx1+stx2 positive, and 6.9% (n = 9) harbored stx1 alone. The stx subtype most frequently detected was stx2a (54.6%, n = 71), followed by stx1a (26.9%, n = 35), stx2c (22.3%, n = 29), stx2d (16.2%, n = 21), stx2b and stx2g (1.5%, n = 2), and stx1c (0.8%, n = 1). Among the other virulence genes assessed, the most frequently detected was ehaA (96.2%, n = 125), followed by iha (91.5%, n = 119), lpfA (77.7%, n = 101), ehxA (60%, n = 78), hlyA (58.5%, n = 76), tia (41.5%, n = 54), saa (40.8%, n = 53), subA (32.3%, n = 42), eae (30.8%, n = 40), cdtB (20%, n = 26), and sab (17.7%, n = 23). Additionally, 43.1% (n = 56) harbored LAA, with its associated genes pagC-like and hes (83.9%, n = 47), ag43 (64.3%, n = 36), and lesP (28.6%, n = 16).
Studies focusing on the AMR of STEC strains isolated from the livestock-beef-human interface in South America are scarce [38,41,61,62,63]. In this study, genomic analysis revealed a low prevalence of resistance genes. In this regard, some strains carried aminoglycoside phosphotransferases encoding genes aph(3″)-Ib (3.1%, n = 4), aph(6)-Id (2.3%, n = 3), and aph(3′)-Ia (0.8%, n = 1), which reduce the electrostatic interaction of the drug and the bacterial rRNA [64]. Four strains harbored tetA or tetB genes, which encodes for tetracycline efflux proteins [65]. Three strains harbored blaTEM-1 β-lactamase encoding genes, which hydrolyze penicillins and narrow-spectrum cephalosporins [66]. Two strains harbored the sul2 gene, which encodes a drug-resistant dihydropteroate synthase, reducing affinity with sulfonamides [67]. Two strains harbored the fosA7 gene, encoding a fosfomycin inactivating enzyme [68], and two strains harbored the qnrB19 gene, which encodes the Qnr protein that interacts with GyrA/B, preventing the action of fluoroquinolones [69]. As expected, all strains harbored chromosomal blaAmpC [70] (Table S2).

3.3. Sequence Types, Serotypes and Phylogenomic Analysis of STEC Strains Circulating in South America

MLST typing revealed the presence of 41 STs among the circulating STEC strains, most of them belonging to ST11 (CC11) (20.8%, n = 27), followed by ST297 (9.2%, n = 12), and ST58 (CC155) (5.4%, n = 7). Figure 1 shows the distribution of the STs of STEC strains circulating in South America, based on analysis of publicly available genomes and data obtained from previous surveillance studies [21,39,52,53,54,55,56,61]. Of clinical interest, ST677, ST58, ST16, ST342, and ST11 carried multiple resistance genes. In addition to blaAmpC, STEC strains of ST677 and ST58 isolated from beef samples in Chile carried aph(3″)-Ib, aph(6)-Id and tetB (n = 2), aph(3″)-Ib, aph(6)-Id, sul2 and tetB (n = 1), or qnrB19 (n = 2) resistance genes; whereas human STEC strains belonging to ST16 and ST342, from Uruguay, carried aph(3′)-Ia, aph(3″)-Ib, blaAmpC, blaTEM-1B, sul2 and tetA (n = 1), or blaAmpC and fosA7 (n = 2) resistance genes, respectively. Two human STEC belonging to ST11, from Paraguay, carried blaAmpC and blaTEM-1B genes.
Among the 130 STEC genomes analyzed, the most frequently detected serotypes were O-:H7 (10.8%, n = 14) and O157:H7 (10%, n = 13), followed by O113:H21 (9.2%, n = 12). Table S2 summarizes the STs, CCs, and serotypes from all STEC genomes analyzed.
We further investigated the genomic relatedness among STEC lineages identified in this study with other STEC strains isolated in South America from human, cattle, and beef samples. The minimum spanning tree of the wgMLST analysis revealed that the 21 STEC strains sequenced in this study (genome IDs in red letters in Figure 2), clustered with other STEC strains previously isolated from cattle or beef in Chile, supporting the persistence of some clones circulating within the country, such as ST332, ST297, ST443, and ST1125 (Figure 2). On the other hand, some clusters included STEC of ST679 from beef (Uruguay) and human (Argentina), ST223 from beef and cattle (Chile), ST21 from human (Argentina) and cattle (Chile), ST332 from cattle (Chile) and beef (Chile and Argentina), and ST443 from beef (Uruguay) and cattle (Chile). For ST11, two epidemiologically relevant clusters were identified. The first included human strains from Argentina, Uruguay, and Paraguay, and the second cluster included STEC strains from human samples collected in Argentina and Paraguay and beef samples collected in Argentina (Figure 2).
SNP-based phylogenetic analysis confirmed genomic relatedness between STEC strains belonging to ST448/O148:H8 (B-18, B-19, and B-20, difference ≤ 246 SNPs), ST297/O130:H11 (B-32, B-40, and RA-13, difference ≤ 339 SNPs, ST1125/-O:H19 (B-6, B-58, and B-60, difference ≤ 238 SNPs), ST657/O183:h18 (B-61) and ST11238/O183:H18 (B-64) with difference of 124 SNPs, and ST33/O91:H14 and novel ST(-1661)/O91:H14 (B-13, B-15, and B-16, difference ≤ 431 SNPs). Additionally, most clonally related STs shared an identical virulome and resistome (Figure 3 and Table S3).

3.4. Association Between stx Type and the Geographic Location and Isolation Source of STEC Strains

The logistic regression model for stx1 positivity showed a significant association with the isolation source; stx2 showed no significant association with any of the included variables, while for both stx1 and stx2 positivity showed a significant association with the geographic location of the sample (Table 1).

4. Discussion

Internationally, several studies have revealed a broad diversity of genomic patterns among STEC strains [32,47,71,72,73]. As far as we know, the present study is the first to analyze genomic patterns of STEC strains circulating in South America. Strains of the O157:H7 serotype have been shown to be genotypically diverse [74], with nine different clades identified by phylogenetic analysis [75]. Clades 6 and 8 are more frequently associated with severe disease in humans and with host-associated fitness [74,76,77]. However, this distribution may vary according to their geographic origin [78,79,80], suggesting a divergent evolution. On the other hand, non-O157 STEC strains show a broader phylogenomic diversity with significant differences in virulotypes [12,81,82]. Phylogenomic analysis corroborated these findings, where all O157 strains clustered together in the same clade, whereas non-O157 strains showed high diversity. All O-:H7 strains clustered together with the O157:H7 strains. This may have been caused by the horizontal transfer and replacement of a part or all of the O-antigen biosynthesis gene cluster from an original O157:H7 strain [31,83].
Serotypes O157:H7 and O113:H21 have been the most frequently detected in cattle, beef, other foodstuffs, and humans in South American countries [19,55,61,84,85,86]. In silico analysis confirmed that 96.3% of the O157:H7 strains were isolated in Argentina from beef and human clinical samples, while all O113:H21 strains were isolated in Chile from beef and cattle samples, suggesting selection and predominance of specific serotypes within each country.
ST11 (CC11), ST58 (155), and ST297 were the most frequently detected. Those STs were isolated worldwide from human and non-human hosts, and are associated with illness both in humans and animals [47,87]. While ST11 is the main clone identified from human infections in Argentina, Brazil, Uruguay, and Paraguay; lineages of CC29 (ST16 and ST21) have emerged in human infections in Argentina, Ecuador, Brazil, Uruguay, and Chile; the latter being further identified in cattle [21,39,52,53,54,55]. Further epidemiologically important data observed from MLST analysis was the successful expansion of the ST11/O157:H7 clone through Brazil, Uruguay, Paraguay, and Argentina, where it has caused severe human infections, including HUS [21]. In the same way, non-O157 STEC lineages of the CC29 (ST16 (O111:H8) and ST21 (O26:H11)) have been isolated from human infections in Argentina, Brazil, Uruguay, and Ecuador; where in Chile this clone was identified in cattle. Specifically, ST21 (O26:H11) strains have been recognized as an emerging HUS-associated STEC lineage in European and North American countries [88,89], where ST16 has been isolated from cases reporting recent travel abroad in England [47,89].
Some specific STs have been identified in two or three countries suggesting common activities, including multilateral trade and travel. The international STEC clone ST223/CC155 (O113:H21) was identified in cattle and beef in Chile (this study) and it has been previously identified in cattle, beef, and hamburgers in Argentina [55], and in cattle in Brazil [53]. In fact, ST223 has been isolated from environment, food, and clinical infections from various European countries as well [53,90]. Worryingly, in Argentina and Thailand, this clone has been associated with HUS [53].
STEC strains of the ST58 have been present in Chile (cattle) and Brazil (humans) [21,61], and have been isolated from cattle in Sweden [90]. Strikingly, as well as the ST223 (alleles adk-6, fumC-4, gyrB-4, icd-18, mdh-24, purA-8, recA-14), the ST58 (alleles adk-6, fumC-4, gyrB-4, icd-16, mdh-24, purA-8, recA-14) is a single-locus variant of the ST155 (alleles adk-6, fumC-4, gyrB-14, icd-16, mdh-24, purA-8, recA-14), which has been designated as the primary founder of the CC155. The ST prediction performed in this study identified the presence of the ST677 (O174:H21) clone in beef samples from Chile and Argentina, whereas ST443 (O178:H19) and ST2388 (O15:H27) were identified in cattle and beef samples from Chile and Uruguay. While ST679 (O163:H19) was predicted in beef and human STEC genomes from Uruguay and Argentina, respectively, previous studies have reported the presence of ST78 and ST106 in Peruvian children and Argentinean cattle [52,55]. In this regard, ST78, a common ST in clinical strains, has been a predominant STEC genotype among cattle and environmental strains [91]. ST101 and S297 were also predicted from genomes of STEC strains isolated from humans and beef in Brazil and Chile, respectively. On the other hand, while ST101 has been identified in food samples in South Korea [92], the ST297 has been a predominant lineage in cattle from Sweden [90], and in pigs, cattle, milk, and water from dairy farms in China [93].
Interestingly, the phylogenetic analysis performed to evaluate the genomic relatedness among STEC strains confirmed the presence of clusters that include clones circulating at the livestock-food-human interface, supporting the persistence, adaptation, and successful expansion of specific lineages across and within different countries.
The most detected stx type was stx2, which has been shown to be associated with more virulent STEC lineages and HUS. In fact, Stx2 presents an LD50 400 times lower than Stx1 in murine models [94]. Among the Stx subtypes, Stx2a, Stx2b, Stx2c, or Stx2d, have been most frequently associated with the development of HUS in infected patients. Other Stx subtypes have been associated with mild illness in humans without complications or as asymptomatic carriers [3,95]. Additionally, Stx2a is epidemiologically associated with increased excretion levels of STEC O157 from cattle [96] and increased transmission between animals, presumably because it is more rapidly produced than other Stx subtypes and restricts cellular proliferation of bovine epithelial cells [97].
Another important virulence factor of STEC is the eae gene, which has traditionally been recognized as a virulence-marker, but here only 30.8% of strains harbored this gene. These eae-positive strains included ST11 lineages, the “big six” (O26:H11, O103:H2, O111:H8, O145:H25, and O145:H28) and other serotypes (O69:H11, O98:H21, O172:H25) considered emergent [98]. However, the burden of illness caused by LEE-negative STEC strains has recently increased in several countries, such as Argentina, Chile, and Paraguay [99]. In this study, most of the analyzed strains were LEE-negative, but harbored different adhesin-encoding genes, including lpfA, ehaA, and saa. LpfA correspond to a major fimbrial subunit protein that is able to interact with fibronectin, laminin, and collagen IV [100]; EhaA is an autotransporter protein related with biofilm and cellular aggregation; while Saa is considered an LEE-negative marker [48] increasing adhesion to HEp-2 cells nearly 10-fold [101]. Moreover, some recently acquired pathogenicity islands (PAI) could contribute to its adhesion, such as LAA. Those PAI can be present as a complete structure with their four modules: module I (hes and other genes), module II (iha, lesP, and others), module III (pagC-like and other genes), and module IV (ag43 and other genes), or as an incomplete structure with some missing modules. Among analyzed strains, this PAI was identified in 43.1% of the strains, mostly as a complete structure. The acquisition of this PAI is probably a recent evolutionary event in STEC [35], which could have contributed to the emergence of highly virulent LEE-negative strains associated with HC and HUS, which are widely distributed in South American countries [25,52,102,103,104].
Other toxin-encoding genes detected were ehxA, hlyA, subA, and cdtB, these last two only being detected in LEE-negative strains. The α-hemolysin (HlyA) and the plasmid-encoded enterohemolysin (EhxA) are widely distributed in STEC strains and have been frequently associated with mild to severe illness in humans [105,106]. CDT causes irreversible G2/M arrest, inhibition of proliferation, and death of human endothelial cells and is frequently detected in LEE-negative strains associated with HUS [107]. SubAB is highly toxic for a range of cell types and induces vacuolization and temporarily protein synthesis inhibition [108], and has a synergic effect with Stx2 in human glomerular endothelial cells damage, contributing to the development of HUS [109]. Given these results, the STEC strains circulating in South America, despite their geographical origin and isolation source, pose a public health risk.
Although antibiotic treatment of STEC infections in humans is not recommended, as they may worsen the disease by inducing toxin-related tissue damage and symptoms in patients [1], STEC can easily exchange AMR encoding genes with different bacterial species, within their hosts and in the environment [110]. STEC strains resistant to β-lactams, aminoglycosides, fluoroquinolones, phenicols, and tetracyclines, among others, have been isolated worldwide from livestock, beef, and humans [36,111,112,113]. Despite the low detection of AMR genes registered in this study, most identified genes are associated resistance to critically important or highly important veterinary and human antibiotics [114,115]; representing a critical issue. This low content of resistance genes could be the result of national legislation concerning antimicrobial use in livestock. For example, in Chile the use of any kind of antimicrobials as growth promoters has been banned since 2006 [116], and as prophylactics since 2019 [117], whereas in Uruguay antimicrobial use for the same purpose has been prohibited in cattle and sheep feed since 2011 [118]. Nevertheless, to date, the use of some antibacterial agents as growth promoters in livestock are still permitted in Argentina and Brazil.

5. Conclusions

We report the genomic characteristics of STEC strains circulating in the livestock-beef-human interface in South America, highlighting the successful expansion of O157:H7 (ST11) and non-O157 (ST16/CC29-O111:H8, ST21/CC29-O26:H11, ST223/CC155-O113:H21, ST58/CC155, ST677-O174:H21, ST443 -O178:H19, ST2388-O15:H27, ST679-O163:H19, ST78, ST106, ST101, and ST297) clones, most likely favored by common activities, such as multilateral trade and travel. Worryingly, some of these STEC clones have been isolated from severe human infections, including HUS, representing a risk for food safety and public health. The logistic regression model indicated that STEC isolates from cattle are more likely to harbor stx1 than those isolated from beef, and that stx2 seems to be a common feature of STEC strains isolated from cattle, beef products, and humans in South America. Additionally, STEC strains from South American countries other than Argentina and Chile present a higher probability of carrying both types of stx. It is important to establish that this study was performed using publicly available genome sequences, which could represent a bias due to underreporting of STEC isolates in this geographical region, highlighting the importance of establishing integrated surveillance programs, both national and regional, ensuring a One Health approach. In summary, these findings support the need for continuous monitoring and surveillance of STEC strains in South America, not only focusing on the detection of O157:H7 serotypes, but also on resistance profiles, virulome, and STs. In this regard, genomic surveillance can be used to rapidly identify and prevent the spread of critical clones “from farm to table”.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ani11071845/s1. Table S1: Name, Sequence Read Archive (SRA) run accession numbers, collection information and ID species results for all 130 STEC strains isolated from cattle, beef and humans analyzed in this study. Table S2: Source, origin, isolation year, phylogroup, sequence type (ST), clonal complex (CC), serotype, virulome, AMR genes of 130 STEC strains isolated from cattle, beef and humans in South America deposited at GenBank. Table S3: SNP matrix STEC Chile.

Author Contributions

Conceptualization, N.G.; methodology, N.G. and F.S.; software, F.S., V.M., D.F.-C., T.J. and R.A.-M.; validation, F.S., V.M., D.F.-C.,T.J. and R.A.-M.; formal analysis, F.S., V.M., D.F.-C.,T.J., E.S. and R.A.-M.; investigation, N.G., B.E., N.L. and V.M.; resources, N.G., V.N., T.J. and N.L.; data curation, N.G., F.S., V.M. and N.L.; writing—original draft preparation, N.G.; writing—review and editing, N.G., L.L., J.C., V.M. and N.L.; visualization, N.G., L.L., J.C., V.M. and N.L.; supervision, N.G.; project administration, N.G.; funding acquisition, N.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Fondo Nacional de Desarrollo Científico y Tecnológico (FONDECYT) grant number 11170363. F.S. is scholar of the ANID/Scholarship Program/Beca de Doctorado Nacional/2021-21210132, and N.L. is a research fellow of the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq grant 312249/2017-9).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Given that in this study only bacterial strains and STEC whole genome sequences were used, no ethical permissions were required. Whole-genome sequences of STEC strains sequenced here are deposited in GenBank under BioProject numbers PRJNA656305 and PRJNA682583.

Acknowledgments

We would like to thank Consuelo Borie for her kind contribution in the development and revision of this study, and to the Molecular Diagnostic and Development laboratory staff at Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, for sample processing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Melton-Celsa, A.R. Shiga Toxin (Stx) Classification, structure, and function. Microbiol. Spectr. 2014, 2, 2–4. [Google Scholar] [CrossRef] [Green Version]
  2. Gyles, C.L.; Fairbrother, J.M. Escherichia coli. In Pathogenesis of Bacterial Infections in Animals; John Wiley & Sons: Hoboken, NJ, USA, 2010; pp. 267–308. ISBN 978-0-470-95820-9. [Google Scholar]
  3. Amézquita-López, B.A.; Soto-Beltrán, M.; Lee, B.G.; Yambao, J.C.; Quiñones, B. Isolation, genotyping and antimicrobial resistance of Shiga toxin-producing Escherichia coli. J. Microbiol. Immunol. Infect. 2018, 51, 425–434. [Google Scholar] [CrossRef] [PubMed]
  4. Sapountzis, P.; Segura, A.; Desvaux, M.; Forano, E. An overview of the elusive passenger in the gastrointestinal tract of cattle: The Shiga toxin producing Escherichia coli. Microorganisms 2020, 8, 877. [Google Scholar] [CrossRef] [PubMed]
  5. Rivas, M.; Chinen, I.; Guth, B.E.C. Enterohemorrhagic (Shiga toxin-producing) Escherichia coli. In Escherichia coli in the Americas; Torres, A.G., Ed.; Springer International Publishing: Manhattan, NY, USA, 2016; pp. 97–123. ISBN 978-3-319-45092-6. [Google Scholar]
  6. Fernández, D.; Sanz, M.E.; Parma, A.E.; Padola, N.L. Short Communication: Characterization of Shiga toxin-producing Escherichia coli isolated from newborn, milk-fed, and growing calves in Argentina. J. Dairy Sci. 2012, 95, 5340–5343. [Google Scholar] [CrossRef]
  7. Rivera, F.P.; Sotelo, E.; Morales, I.; Menacho, F.; Medina, A.M.; Evaristo, R.; Valencia, R.; Carbajal, L.; Ruiz, J.; Ochoa, T.J. Short communication: Detection of Shiga toxin-producing Escherichia coli (STEC) in healthy cattle and pigs in Lima, Peru. J. Dairy Sci. 2012, 95, 1166–1169. [Google Scholar] [CrossRef] [PubMed]
  8. Galarce, N.; Escobar, B.; Sánchez, F.; Paredes-Osses, E.; Alegría-Morán, R.; Borie, C. Virulence genes, Shiga toxin subtypes, serogroups, and clonal relationship of Shiga toxin-producing Escherichia coli strains isolated from livestock and companion animals. Animals 2019, 9, 733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Gonzalez, A.G.M.; Cerqueira, A.M.F. Shiga Toxin-producing Escherichia coli in the animal reservoir and food in Brazil. J. Appl. Microbiol. 2020, 128, 1568–1582. [Google Scholar] [CrossRef] [Green Version]
  10. Smith, J.L.; Fratamico, P.M. Emerging and re-emerging foodborne pathogens. Foodborne Pathog. Dis. 2018, 15, 737–757. [Google Scholar] [CrossRef]
  11. Scott, M.E.; Mbandi, E.; Buchanan, S.; Abdelmajid, N.; Gonzalez-Rivera, C.; Hale, K.R.; Jacobsen, L.; Webb, J.; Green, J.; Dolan, P. Salmonella and Shiga toxin–producing Escherichia coli in products sampled in the food safety and inspection service raw pork baseline study. J. Food Protect. 2020, 83, 552–559. [Google Scholar] [CrossRef]
  12. Franz, E.; Delaquis, P.; Morabito, S.; Beutin, L.; Gobius, K.; Rasko, D.A.; Bono, J.; French, N.; Osek, J.; Lindstedt, B.-A.; et al. Exploiting the explosion of information associated with whole genome sequencing to tackle Shiga toxin-producing Escherichia coli (STEC) in global food production systems. Int. J. Food Microbiol. 2014, 187, 57–72. [Google Scholar] [CrossRef]
  13. Food Safety and Inspection Service, Department of Agriculture, USA (FSIS). Shiga Toxin-Producing Escherichia coli in Certain Raw Beef Products. Available online: https://www.govinfo.gov/content/pkg/FR-2011-11-23/pdf/2011-30271.pdf (accessed on 31 March 2021).
  14. Shakerian, A.; Rahimi, E.; Emad, P. Vegetables and restaurant salads as a reservoir for Shiga toxigenic Escherichia coli: Distribution of virulence factors, O-serogroups, and antibiotic resistance properties. J. Food Prot. 2016, 79, 1154–1160. [Google Scholar] [CrossRef]
  15. Abe, C.M.; Matheus-Guimarães, C.; Garcia, B.G.; Cabilio Guth, B.E. Interactions of Shiga toxin-producing Escherichia coli with leafy green vegetables. Braz. J. Microbiol. 2020, 51, 797–803. [Google Scholar] [CrossRef] [PubMed]
  16. Cody, E.M.; Dixon, B.P. Hemolytic uremic syndrome. Pediatr. Clin. N. Am. 2019, 66, 235–246. [Google Scholar] [CrossRef] [PubMed]
  17. Majowicz, S.E.; Scallan, E.; Jones-Bitton, A.; Sargeant, J.M.; Stapleton, J.; Angulo, F.J.; Yeung, D.H.; Kirk, M.D. Global Incidence of human Shiga toxin-producing Escherichia coli infections and deaths: A systematic review and knowledge synthesis. Foodborne Pathog. Dis. 2014, 11, 447–455. [Google Scholar] [CrossRef] [Green Version]
  18. Food and Agriculture Organization of the United Nations (FAO); World Health Organization (WHO). Attributing Illness Caused by Shiga Toxin-Producing Escherichia coli (STEC) to Specific Foods; Microbiological Risk Assessment Series; FAO-WHO: Rome, Italy, 2019; ISBN 978-92-5-131746-4. [Google Scholar]
  19. Torres, A.G.; Amaral, M.M.; Bentancor, L.; Galli, L.; Goldstein, J.; Krüger, A.; Rojas-Lopez, M. Recent advances in Shiga toxin-producing Escherichia coli research in Latin America. Microorganisms 2018, 6, 100. [Google Scholar] [CrossRef] [Green Version]
  20. Castro, V.S.; Teixeira, L.A.C.; Rodrigues, D.D.P.; Dos Santos, L.F.; Conte-Junior, C.A.; de Souza Figueiredo, E.E. Occurrence and antimicrobial resistance of E. coli non-O157 isolated from beef in Mato Grosso, Brazil. Trop. Anim. Health Prod. 2019, 51, 1117–1123. [Google Scholar] [CrossRef]
  21. Cavalcanti, A.M.F.; Hernandes, R.T.; Takagi, E.H.; Guth, B.E.C.; de Lima Ori, É.; Pinheiro, S.R.S.; de Andrade, T.S.; Oliveira, S.L.; Cergole-Novella, M.C.; Francisco, G.R.; et al. Virulence profiling and molecular typing of Shiga toxin-producing E. coli (STEC) from human sources in Brazil. Microorganisms 2020, 8, 171. [Google Scholar] [CrossRef] [Green Version]
  22. Organization for Economic Co-Operation and Development (OECD). Food and Agriculture Organization of the United Nations (FAO) Meat. In OECD-FAO Agricultural Outlook 2020–2029; OECD-FAO: Rome, Italy, 2020; Available online: http://www.fao.org/3/ca8861en/Meat.pdf (accessed on 31 March 2021).
  23. Food and Agriculture Organization of the United Nations Statistics Division (FAOSTAT). Datos Sobre Alimentación y Agricultura. Available online: http://www.fao.org/faostat/es/#home (accessed on 31 March 2021).
  24. Pianciola, L.; Rivas, M. Genotypic features of clinical and bovine Escherichia coli O157 strains isolated in countries with different associated-disease incidences. Microorganisms 2018, 6, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Pianciola, L.; D’Astek, B.A.; Mazzeo, M.; Chinen, I.; Masana, M.; Rivas, M. Genetic features of human and bovine Escherichia coli O157:H7 strains isolated in Argentina. Int. J. Med. Microbiol. 2016, 306, 123–130. [Google Scholar] [CrossRef] [PubMed]
  26. Castro, V.S.; Carvalho, R.C.T.; Conte-Junior, C.A.; Figuiredo, E.E.S. Shiga-toxin producing Escherichia coli: Pathogenicity, supershedding, diagnostic methods, occurrence, and foodborne outbreaks. Comp. Rev. Food Sci. Food Saf. 2017, 16, 1269–1280. [Google Scholar] [CrossRef] [Green Version]
  27. Borie, C.F.; Monreal, Z.; Martinez, J.; Arellano, C.; Prado, V. Detection and characterization of Enterohaemorrhagic Escherichia coli in slaughtered cattle. Zent. Vet. B 1997, 44, 273–279. [Google Scholar] [CrossRef]
  28. Blanco, M.; Padola, N.L.; Krüger, A.; Sanz, M.E.; Blanco, J.E.; González, E.A.; Dahbi, G.; Mora, A.; Bernárdez, M.I.; Etcheverría, A.I.; et al. Virulence genes and intimin types of Shiga-toxin-producing Escherichia coli isolated from cattle and beef products in Argentina. Int. Microbiol. 2004, 7, 269–276. [Google Scholar] [PubMed]
  29. Pigatto, C.P.; Schocken-Iturrino, R.P.; Souza, E.M.; Pedrosa, F.O.; Comarella, L.; Irino, K.; Kato, M.A.M.F.; Farah, S.M.S.S.; Warth, J.F.; Fadel-Picheth, C.M.T. Virulence properties and antimicrobial susceptibility of Shiga toxin-producing Escherichia coli strains isolated from healthy cattle from Paraná State, Brazil. Can. J. Microbiol. 2008, 54, 588–593. [Google Scholar] [CrossRef]
  30. Fernández, D.; Irino, K.; Sanz, M.E.; Padola, N.L.; Parma, A.E. Characterization of Shiga toxin-producing Escherichia coli isolated from dairy cows in Argentina. Lett. Appl. Microbiol. 2010, 51, 377–382. [Google Scholar] [CrossRef]
  31. Iguchi, A.; Shirai, H.; Seto, K.; Ooka, T.; Ogura, Y.; Hayashi, T.; Osawa, K.; Osawa, R. Wide distribution of O157-antigen biosynthesis gene clusters in Escherichia coli. PLoS ONE 2011, 6, e23250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ferdous, M.; Friedrich, A.W.; Grundmann, H.; de Boer, R.F.; Croughs, P.D.; Islam, M.A.; Kluytmans-van den Bergh, M.F.Q.; Kooistra-Smid, A.M.D.; Rossen, J.W.A. Molecular characterization and phylogeny of Shiga toxin-producing Escherichia coli isolates obtained from two Dutch regions using whole genome sequencing. Clin. Microbiol. Infect. 2016, 22, 642.e1. [Google Scholar] [CrossRef] [Green Version]
  33. Koutsoumanis, K.; Allende, A.; Alvarez-Ordóñez, A.; Bover-Cid, S.; Chemaly, M.; Davies, R.; Cesare, A.D.; Herman, L.; Hilbert, F.; Lindqvist, R.; et al. Pathogenicity assessment of Shiga toxin-producing Escherichia coli (STEC) and the public health risk posed by contamination of food with STEC. EFSA J. 2020, 18, e05967. [Google Scholar] [CrossRef]
  34. Paton, A.W.; Srimanote, P.; Woodrow, M.C.; Paton, J.C. Characterization of Saa, a novel autoagglutinating adhesin produced by Locus of Enterocyte Effacement-negative Shiga-toxigenic Escherichia coli strains that are virulent for humans. Infect. Immun. 2001, 69, 6999–7009. [Google Scholar] [CrossRef] [Green Version]
  35. Montero, D.A.; Velasco, J.; Del Canto, F.; Puente, J.L.; Padola, N.L.; Rasko, D.A.; Farfán, M.; Salazar, J.C.; Vidal, R. Locus of Adhesion and Autoaggregation (LAA), a pathogenicity island present in emerging Shiga toxin–producing Escherichia coli strains. Sci. Rep. 2017, 7, 7011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ahmed, A.M.; Shimamoto, T. Molecular analysis of multidrug resistance in Shiga toxin-producing Escherichia coli O157: H7 isolated from meat and dairy products. Int. J. Food Microbiol. 2015, 193, 68–73. [Google Scholar] [CrossRef] [PubMed]
  37. Park, H.-J.; Yoon, J.W.; Heo, E.-J.; Ko, E.-K.; Kim, K.-Y.; Kim, Y.-J.; Yoon, H.-J.; Wee, S.-H.; Park, Y.H.; Moon, J.S. Antibiotic resistance and virulence potentials of Shiga toxin-producing Escherichia coli isolates from raw meats of slaughterhouses and retail markets in Korea. J. Microbiol. Biotechnol. 2015, 25, 1460–1466. [Google Scholar] [CrossRef]
  38. Colello, R.; Krüger, A.; Conza, J.D.; Rossen, J.W.A.; Friedrich, A.W.; Gutkind, G.; Etcheverría, A.I.; Padola, N.L. Antimicrobial resistance in class 1 integron-positive Shiga toxin-producing Escherichia coli isolated from cattle, pigs, food and farm environment. Microorganisms 2018, 6, 99. [Google Scholar] [CrossRef] [Green Version]
  39. Furlan, J.P.R.; Gallo, I.F.L.; de Campos, A.C.L.P.; Passaglia, J.; Falcão, J.P.; Navarro, A.; Nakazato, G.; Stehling, E.G. Molecular characterization of multidrug-resistant Shiga toxin-producing Escherichia coli harboring antimicrobial resistance genes obtained from a farmhouse. Pathog. Glob. Health 2019, 113, 268–274. [Google Scholar] [CrossRef]
  40. Puii, L.H.; Dutta, T.K.; Roychoudhury, P.; Kylla, H.; Chakraborty, S.; Mandakini, R.; Kawlni, L.; Samanta, I.; Bandopaddhay, S.; Singh, S.B. Extended spectrum beta-lactamase producing Shiga-toxin producing-Escherichia coli in piglets, humans and water sources in North East Region of India. Lett. Appl. Microbiol. 2019, 69, 373–378. [Google Scholar] [CrossRef] [PubMed]
  41. Galarce, N.; Sánchez, F.; Fuenzalida, V.; Ramos, R.; Escobar, B.; Lapierre, L.; Paredes-Osses, E.; Arriagada, G.; Alegría-Morán, R.; Lincopán, N.; et al. Phenotypic and genotypic antimicrobial resistance in non-O157 Shiga toxin-producing Escherichia coli isolated from cattle and swine in Chile. Front. Vet. Sci. 2020, 7, 367. [Google Scholar] [CrossRef] [PubMed]
  42. Dhanji, H.; Murphy, N.M.; Doumith, M.; Durmus, S.; Lee, S.S.; Hope, R.; Woodford, N.; Livermore, D.M. Cephalosporin resistance mechanisms in Escherichia coli isolated from raw chicken imported into the UK. J. Antimicrob. Chemother. 2010, 65, 2534–2537. [Google Scholar] [CrossRef] [Green Version]
  43. Alonso, C.A.; Zarazaga, M.; Sallem, R.B.; Jouini, A.; Slama, K.B.; Torres, C. Antibiotic resistance in Escherichia coli in husbandry animals: The African perspective. Lett. Appl. Microbiol. 2017, 64, 318–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Kim, J.-S.; Lee, M.-S.; Kim, J.H. Recent updates on outbreaks of Shiga toxin-producing Escherichia coli and its potential reservoirs. Front. Cell Infect. Microbiol. 2020, 10, 273. [Google Scholar] [CrossRef]
  45. Dallman, T.J.; Byrne, L.; Ashton, P.M.; Cowley, L.A.; Perry, N.T.; Adak, G.; Petrovska, L.; Ellis, R.J.; Elson, R.; Underwood, A.; et al. Whole-genome sequencing for national surveillance of Shiga toxin-producing Escherichia coli O157. Clin. Infect. Dis. 2015, 61, 305–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Baba, H.; Kanamori, H.; Kudo, H.; Kuroki, Y.; Higashi, S.; Oka, K.; Takahashi, M.; Yoshida, M.; Oshima, K.; Aoyagi, T.; et al. Genomic analysis of Shiga toxin-producing Escherichia coli from patients and asymptomatic food handlers in Japan. PLoS ONE 2019, 14, e0225340. [Google Scholar] [CrossRef]
  47. González-Escalona, N.; Kase, J.A. Virulence gene profiles and phylogeny of Shiga toxin-positive Escherichia coli strains isolated from FDA regulated foods during 2010–2017. PLoS ONE 2019, 14, e0214620. [Google Scholar] [CrossRef] [Green Version]
  48. Montero, D.A.; Canto, F.D.; Velasco, J.; Colello, R.; Padola, N.L.; Salazar, J.C.; Martin, C.S.; Oñate, A.; Blanco, J.; Rasko, D.A.; et al. Cumulative acquisition of pathogenicity islands has shaped virulence potential and contributed to the emergence of LEE-negative Shiga toxin-producing Escherichia coli strains. Emerg. Microbes. Infect. 2019, 8, 486–502. [Google Scholar] [CrossRef] [Green Version]
  49. Pavez-Muñoz, E.; González, C.; Fernández-Sanhueza, B.; Sánchez, F.; Escobar, B.; Ramos, R.; Fuenzalida, V.; Galarce, N.; Arriagada, G.; Neira, V.; et al. Antimicrobial usage factors and resistance profiles of Shiga toxin-producing Escherichia coli in backyard production systems from Central Chile. Front. Vet. Sci. 2020, 7, 595149. [Google Scholar] [CrossRef]
  50. Zhou, Z.; Alikhan, N.-F.; Mohamed, K.; Fan, Y.; Achtman, M.; Agama Study Group. The EnteroBase user’s guide, with case studies on Salmonella transmissions, Yersinia pestis phylogeny, and Escherichia core genomic diversity. Genome Res. 2020, 30, 138–152. [Google Scholar] [CrossRef] [Green Version]
  51. Li, H.; Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 2009, 25, 1754–1760. [Google Scholar] [CrossRef] [Green Version]
  52. Contreras, C.A.; Ochoa, T.J.; Ruiz, J.; Lacher, D.W.; Rivera, F.P.; Saenz, Y.; Chea-Woo, E.; Zavaleta, N.; Gil, A.I.; Lanata, C.F.; et al. Phylogenetic relationships of Shiga toxin-producing Escherichia coli isolated from Peruvian Children. J. Med. Microbiol. 2011, 60, 639–646. [Google Scholar] [CrossRef] [Green Version]
  53. Feng, P.C.H.; Delannoy, S.; Lacher, D.W.; Dos Santos, L.F.; Beutin, L.; Fach, P.; Rivas, M.; Hartland, E.L.; Paton, A.W.; Guth, B.E.C. Genetic diversity and virulence potential of Shiga toxin-producing Escherichia coli O113:H21 strains isolated from clinical, environmental, and food sources. Appl. Environ. Microbiol. 2014, 80, 4757–4763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Amézquita-Montes, Z.; Tamborski, M.; Kopsombut, U.G.; Zhang, C.; Arzuza, O.S.; Gómez-Duarte, O.G. Genetic relatedness among Escherichia coli pathotypes isolated from food products for human consumption in Cartagena, Colombia. Foodborne Pathog. Dis. 2015, 12, 454–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Cadona, J.S.; Bustamante, A.V.; González, J.; Sanso, A.M. Genetic relatedness and novel sequence types of non-O157 Shiga toxin-producing Escherichia coli strains isolated in Argentina. Front. Cell Infect. Microbiol. 2016, 6, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Furlan, J.P.R.; da Silva Ferreira, M.E.; Stehling, E.G. Genetic diversity of multidrug-resistant CMY-producing Escherichia coli from feces and soil in a small-scale pig farm. Microb. Drug Resist. 2020, 26, 1365–1371. [Google Scholar] [CrossRef]
  57. Joensen, K.G.; Tetzschner, A.M.M.; Iguchi, A.; Aarestrup, F.M.; Scheutz, F. Rapid and easy in silico serotyping of Escherichia coli isolates by use of whole-genome sequencing data. J. Clin. Microbiol. 2015, 53, 2410–2426. [Google Scholar] [CrossRef] [Green Version]
  58. Dohoo, I.R.; Martin, S.W.; Stryhn, H. Veterinary Epidemiologic Research; VER: Burbank, CA, USA, 2009; ISBN 978-0-919013-60-5. [Google Scholar]
  59. Hosmer, D.W.; Lemeshow, S.; Sturdivant, R.X. Applied Logistic Regression, 3rd ed.; Wiley Series in Probability and Statistics; Wiley: Hoboken, NJ, USA, 2013; ISBN 978-0-470-58247-3. [Google Scholar]
  60. R Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2013; Available online: https://www.r-project.org/ (accessed on 31 March 2021).
  61. Sánchez, F.; Fuenzalida, V.; Ramos, R.; Escobar, B.; Neira, V.; Borie, C.; Lapierre, L.; López, P.; Venegas, L.; Dettleff, P.; et al. Genomic features and antimicrobial resistance patterns of Shiga toxin-producing Escherichia coli strains isolated from food in Chile. Zoonoses Public Health 2021, 68, 226–238. [Google Scholar] [CrossRef]
  62. Krüger, A.; Lucchesi, P.M.A.; Sanso, A.M.; Etcheverría, A.I.; Bustamante, A.V.; Burgán, J.; Fernández, L.; Fernández, D.; Leotta, G.; Friedrich, A.W.; et al. Genetic characterization of Shiga toxin-producing Escherichia coli O26:H11 strains isolated from animal, food, and clinical samples. Front. Cell Infect. Microbiol. 2015, 5, 74. [Google Scholar] [CrossRef] [Green Version]
  63. Mota, M.I.; Vázquez, S.; Cornejo, C.; D’Alessandro, B.; Braga, V.; Caetano, A.; Betancor, L.; Varela, G. Does Shiga toxin-producing Escherichia coli and Listeria monocytogenes contribute significantly to the burden of antimicrobial resistance in Uruguay? Front. Vet. Sci. 2020, 7, 583930. [Google Scholar] [CrossRef]
  64. Serio, A.W.; Magalhães, M.L.; Blanchard, J.S.; Connolly, L.E. Aminoglycosides: Mechanisms of Action and Resistance. In Antimicrobial Drug Resistance: Mechanisms of Drug Resistance; Mayers, D.L., Sobel, J.D., Ouellette, M., Kaye, K.S., Marchaim, D., Eds.; Springer International Publishing: Manhattan, NY, USA, 2017; Volume 1, pp. 213–229. ISBN 978-3-319-46718-4. [Google Scholar]
  65. Roberts, M.C.; Schwarz, S. Tetracycline and Chloramphenicol Resistance Mechanisms. In Antimicrobial Drug Resistance: Mechanisms of Drug Resistance; Mayers, D.L., Sobel, J.D., Ouellette, M., Kaye, K.S., Marchaim, D., Eds.; Springer International Publishing: Manhattan, NY, USA, 2017; Volume 1, pp. 231–243. ISBN 978-3-319-46718-4. [Google Scholar]
  66. Bush, K. The Importance of β-Lactamases to the Development of New β-Lactams. In Antimicrobial Drug Resistance: Mechanisms of Drug Resistance; Mayers, D.L., Sobel, J.D., Ouellette, M., Kaye, K.S., Marchaim, D., Eds.; Springer International Publishing: Manhattan, NY, USA, 2017; Volume 1, pp. 165–175. ISBN 978-3-319-46718-4. [Google Scholar]
  67. Changkaew, K.; Utrarachkij, F.; Siripanichgon, K.; Nakajima, C.; Suthienkul, O.; Suzuki, Y. Characterization of antibiotic resistance in Escherichia coli isolated from shrimps and their environment. J. Food Prot. 2014, 77, 1394–1401. [Google Scholar] [CrossRef]
  68. Zhanel, G.G.; Walkty, A.J.; Karlowsky, J.A. Fosfomycin: A first-line oral therapy for acute uncomplicated cystitis. Can. J. Infect. Dis. Med. Microbiol. 2016, 2016, 2082693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Schindler, B.D.; Buensalido, J.A.L.; Kaatz, G.W. Fluoroquinolone Resistance in Bacteria. In Antimicrobial Drug Resistance: Mechanisms of Drug Resistance; Mayers, D.L., Sobel, J.D., Ouellette, M., Kaye, K.S., Marchaim, D., Eds.; Springer International Publishing: Manhattan, NY, USA, 2017; Volume 1, pp. 245–263. ISBN 978-3-319-46718-4. [Google Scholar]
  70. Bonomo, R.A. Mutations as a Basis of Antimicrobial Resistance. In Antimicrobial Drug Resistance; Mayers, D.L., Sobel, J.D., Ouellette, M., Kaye, K.S., Marchaim, D., Eds.; Springer International Publishing: Manhattan, NY, USA, 2017; pp. 77–87. ISBN 978-3-319-46716-0. [Google Scholar]
  71. Browne, A.S.; Midwinter, A.C.; Withers, H.; Cookson, A.L.; Biggs, P.J.; Marshall, J.C.; Benschop, J.; Hathaway, S.; Haack, N.A.; Akhter, R.N.; et al. Molecular epidemiology of Shiga toxin-producing Escherichia coli (STEC) on New Zealand dairy farms: Application of a culture-independent assay and whole-genome sequencing. Appl. Environ. Microbiol. 2018, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Abdalhamid, B.; Mccutchen, E.; Bouska, A.; Weiwei, Z.; Loeck, B.; Hinrichs, S.; Iwen, P. Whole genome sequencing to characterize Shiga toxin-producing Escherichia coli O26 in a public health setting. J. Infect. Public Health 2019, 12, 884–889. [Google Scholar] [CrossRef]
  73. Lang, C.; Hiller, M.; Konrad, R.; Fruth, A.; Flieger, A. Whole-genome-based public health surveillance of less common Shiga toxin-producing Escherichia coli serovars and untypeable strains identifies four novel O genotypes. J. Clin. Microbiol. 2019, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Amigo, N.; Mercado, E.; Bentancor, A.; Singh, P.; Vilte, D.; Gerhardt, E.; Zotta, E.; Ibarra, C.; Manning, S.D.; Larzábal, M.; et al. Clade 8 and clade 6 strains of Escherichia coli O157:H7 from cattle in Argentina have hypervirulent-like phenotypes. PLoS ONE 2015, 10, e0127710. [Google Scholar] [CrossRef]
  75. Manning, S.D.; Motiwala, A.S.; Springman, A.C.; Qi, W.; Lacher, D.W.; Ouellette, L.M.; Mladonicky, J.M.; Somsel, P.; Rudrik, J.T.; Dietrich, S.E.; et al. Variation in virulence among clades of Escherichia coli O157: H7 associated with disease outbreaks. Proc. Natl. Acad. Sci. USA 2008, 105, 4868–4873. [Google Scholar] [CrossRef] [Green Version]
  76. Neupane, M.; Abu-Ali, G.; Mitra, A.; Lacher, D.; Manning, S.; Riordan, J. Shiga toxin 2 overexpression in Escherichia coli O157:H7 strains associated with severe human disease. Microb. Pathog. 2011, 51, 466–470. [Google Scholar] [CrossRef] [Green Version]
  77. Kawase, J.; Hirai, S.; Yokoyama, E.; Hayashi, F.; Kurosaki, K.; Kawakami, Y.; Fukuma, A.; Sakai, T.; Kotani, M.; Asakura, H. Phylogeny, prevalence, and Shiga Toxin (Stx) production of clinical Escherichia coli O157 clade 2 strains isolated in Shimane Prefecture, Japan. Curr. Microbiol. 2020, 78, 265–273. [Google Scholar] [CrossRef]
  78. Franz, E.; van Hoek, A.H.A.M.; van der Wal, F.J.; de Boer, A.; Zwartkruis-Nahuis, A.; van der Zwaluw, K.; Aarts, H.J.M.; Heuvelink, A.E. Genetic features differentiating bovine, food, and human isolates of Shiga toxin-producing Escherichia coli O157 in The Netherlands. J. Clin. Microbiol. 2012, 50, 772–780. [Google Scholar] [CrossRef] [Green Version]
  79. Mellor, G.E.; Besser, T.E.; Davis, M.A.; Beavis, B.; Jung, W.; Smith, H.V.; Jennison, A.V.; Doyle, C.J.; Chandry, P.S.; Gobius, K.S.; et al. Multilocus genotype analysis of Escherichia coli O157 isolates from Australia and the United States Provides evidence of geographic divergence. Appl. Environ. Microbiol. 2013, 79, 5050–5058. [Google Scholar] [CrossRef] [Green Version]
  80. Strachan, N.J.C.; Rotariu, O.; Lopes, B.; MacRae, M.; Fairley, S.; Laing, C.; Gannon, V.; Allison, L.J.; Hanson, M.F.; Dallman, T.; et al. Whole genome sequencing demonstrates that geographic variation of Escherichia coli O157 genotypes dominates host association. Sci. Rep. 2015, 5, 14145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Steyert, S.R.; Sahl, J.W.; Fraser, C.M.; Teel, L.D.; Scheutz, F.; Rasko, D.A. Comparative genomics and stx phage characterization of LEE-negative Shiga toxin-producing Escherichia coli. Front. Cell Infect. Microbiol. 2012, 2, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Bumunang, E.W.; McAllister, T.A.; Zaheer, R.; Ortega Polo, R.; Stanford, K.; King, R.; Niu, Y.D.; Ateba, C.N. Characterization of non-O157 Escherichia coli from cattle faecal samples in the North-West Province of South Africa. Microorganisms 2019, 7, 272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Iguchi, A.; Iyoda, S.; Kikuchi, T.; Ogura, Y.; Katsura, K.; Ohnishi, M.; Hayashi, T.; Thomson, N.R. A complete view of the genetic diversity of the Escherichia coli O-antigen biosynthesis gene cluster. DNA Res. 2015, 22, 101–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Miko, A.; Delannoy, S.; Fach, P.; Strockbine, N.A.; Lindstedt, B.A.; Mariani-Kurkdjian, P.; Reetz, J.; Beutin, L. Genotypes and virulence characteristics of Shiga toxin-producing Escherichia coli O104 strains from different origins and sources. Int. J. Med. Microbiol. 2013, 303, 410–421. [Google Scholar] [CrossRef]
  85. Alonso, M.Z.; Krüger, A.; Sanz, M.E.; Padola, N.L.; Lucchesi, P.M.A. Serotypes, virulence profiles and Stx subtypes of Shigatoxigenic Escherichia coli isolated from chicken derived products. Rev. Argent. Microbiol. 2016, 48, 325–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Sanso, A.M.; Bustamante, A.V.; Krüger, A.; Cadona, J.S.; Alfaro, R.; Cáceres, M.E.; Fernández, D.; Lucchesi, P.M.A.; Padola, N.L. Molecular epidemiology of Shiga toxin-producing O113:H21 isolates from cattle and meat. Zoonoses Public Health 2018, 65, 569–577. [Google Scholar] [CrossRef] [PubMed]
  87. Fuentes-Castillo, D.; Navas-Suárez, P.E.; Gondim, M.F.; Esposito, F.; Sacristán, C.; Fontana, H.; Fuga, B.; Piovani, C.; Kooij, R.; Lincopan, N.; et al. Genomic characterization of multidrug-resistant ESBL-producing Escherichia coli ST58 causing fatal colibacillosis in critically endangered Brazilian Merganser (Mergus octosetaceus). Transbound. Emerg. Dis. 2020, 68, 258–266. [Google Scholar] [CrossRef] [PubMed]
  88. Hua, Y.; Chromek, M.; Frykman, A.; Jernberg, C.; Georgieva, V.; Hansson, S.; Zhang, J.; Marits, A.K.; Wan, C.; Matussek, A.; et al. Whole-genome characterization of hemolytic uremic syndrome-causing Shiga toxin-producing Escherichia coli in Sweden. Virulence 2021, 12, 1296–1305. [Google Scholar] [CrossRef]
  89. Dallman, T.J.; Greig, D.R.; Gharbia, S.E.; Jenkins, C. Phylogenetic context of Shiga toxin-producing Escherichia coli serotype O26:H11 in England. Microb. Genom. 2021, 1–9. [Google Scholar] [CrossRef]
  90. Jinnerot, T.; Tomaselli, A.T.P.; Johannessen, G.S.; Söderlund, R.; Urdahl, A.M.; Aspán, A.; Sekse, C. The prevalence and genomic context of Shiga toxin 2a genes in E. coli found in cattle. PLoS ONE 2020, 15, e0232305. [Google Scholar] [CrossRef]
  91. Carter, M.Q.; Quinones, B.; He, X.; Zhong, W.; Louie, J.W.; Lee, B.G.; Yambao, J.C.; Mandrell, R.E.; Cooley, M.B. An environmental Shiga toxin-producing Escherichia coli O145 clonal population exhibits high-level phenotypic variation that includes virulence traits. Appl. Environ. Microbiol. 2016, 82, 1090–1101. [Google Scholar] [CrossRef] [Green Version]
  92. Koo, H.-J.; Kwak, H.-S.; Yoon, S.-H.; Woo, G.-J. Phylogenetic group distribution and prevalence of virulence genes in Escherichia coli isolates from food samples in South Korea. World J. Microbiol. Biotechnol. 2012, 28, 1813–1816. [Google Scholar] [CrossRef]
  93. Peng, Z.; Liang, W.; Hu, Z.; Li, X.; Guo, R.; Hua, L.; Tang, X.; Tan, C.; Chen, H.; Wang, X.; et al. O-serogroups, virulence genes, antimicrobial susceptibility, and MLST genotypes of Shiga toxin-producing Escherichia coli from swine and cattle in Central China. BMC Vet. Res. 2019, 15, 427. [Google Scholar] [CrossRef]
  94. Basu, D.; Tumer, N.E. Do the A subunits contribute to the differences in the toxicity of Shiga toxin 1 and Shiga toxin 2? Toxins 2015, 7, 1467–1485. [Google Scholar] [CrossRef] [Green Version]
  95. Akiyama, Y.; Futai, H.; Saito, E.; Ogita, K.; Sakae, H.; Fukunaga, M.; Tsuji, H.; Chikahira, M.; Iguchi, A. Shiga toxin subtypes and virulence genes in Escherichia coli isolated from cattle. Jpn. J. Infect. Dis. 2017, 70, 181–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Munns, K.D.; Selinger, L.B.; Stanford, K.; Guan, L.; Callaway, T.R.; McAllister, T.A. Perspectives on super-shedding of Escherichia coli O157:H7 by cattle. Foodborne Pathog. Dis. 2015, 12, 89–103. [Google Scholar] [CrossRef]
  97. Fitzgerald, S.F.; Beckett, A.E.; Palarea-Albaladejo, J.; McAteer, S.; Shaaban, S.; Morgan, J.; Ahmad, N.I.; Young, R.; Mabbott, N.A.; Morrison, L.; et al. Shiga toxin sub-type 2a increases the efficiency of Escherichia coli O157 transmission between animals and restricts epithelial regeneration in bovine enteroids. PLoS Pathog. 2019, 15, e1008003. [Google Scholar] [CrossRef] [Green Version]
  98. Sánchez, S.; Llorente, M.T.; Echeita, M.A.; Herrera-León, S. Development of three multiplex PCR assays targeting the 21 most clinically relevant serogroups associated with Shiga toxin-producing E. coli infection in humans. PLoS ONE 2015, 10, e0117660. [Google Scholar] [CrossRef]
  99. Vélez, M.V.; Colello, R.; Etcheverría, A.I.; Vidal, R.M.; Montero, D.A.; Acuña, P.; Guillén Fretes, R.M.; Toro, M.; Padola, N.L. Distribution of Locus of Adhesion and Autoaggregation and hes Gene in STEC strains from countries of Latin America. Curr. Microbiol. 2020, 77, 2111–2117. [Google Scholar] [CrossRef] [PubMed]
  100. Farfan, M.J.; Cantero, L.; Vidal, R.; Botkin, D.J.; Torres, A.G. Long polar fimbriae of Enterohemorrhagic Escherichia coli O157:H7 bind to extracellular matrix proteins. Infect. Immun. 2011, 79, 3744–3750. [Google Scholar] [CrossRef] [Green Version]
  101. McWilliams, B.D.; Torres, A.G. Enterohemorrhagic Escherichia coli adhesins. Microbiol. Spectr. 2014, 2, 131–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Prado, J.V.; Cavagnaro, S.M.F. Síndrome hemolítico urémico asociado a infección intestinal por Escherichia coli productora de Shigatoxina (STEC) en pacientes chilenos: Aspectos clínicos y epidemiológicos. Rev. Chil. Infectol. 2008, 25, 435–444. [Google Scholar] [CrossRef] [Green Version]
  103. de Souza, R.L.; Abreu Carvalhaes, J.T.; Sanae Nishimura, L.; de Andrade, M.C.; Cabilio Guth, B.E. Hemolytic uremic syndrome in pediatric intensive care units in São Paulo, Brazil. Open Microbiol. J. 2011, 5, 76–82. [Google Scholar] [CrossRef]
  104. Pérez, L.; Apezteguía, L.; Piñeyrúa, C.; Dabezies, A.; Bianco, M.N.; Schelotto, F.; Varela, G. Hemolytic uremic syndrome with mild renal involvement due to Shiga toxin-producing Escherichia coli (STEC) O145 strain. Rev. Argent. Microbiol. 2014, 46, 103–106. [Google Scholar] [CrossRef] [Green Version]
  105. Lorenz, S.C.; Son, I.; Maounounen-Laasri, A.; Lin, A.; Fischer, M.; Kase, J.A. Prevalence of hemolysin genes and comparison of ehxA subtype patterns in Shiga toxin-producing Escherichia coli (STEC) and non-STEC strains from clinical, food, and animal sources. Appl. Environ. Microbiol. 2013, 79, 6301–6311. [Google Scholar] [CrossRef] [Green Version]
  106. Fu, S.; Bai, X.; Fan, R.; Sun, H.; Xu, Y.; Xiong, Y. Genetic diversity of the enterohaemolysin gene (ehxa) in non-O157 Shiga toxin-producing Escherichia coli strains in China. Sci. Rep. 2018, 8, 4233. [Google Scholar] [CrossRef]
  107. Bielaszewska, M.; Sinha, B.; Kuczius, T.; Karch, H. Cytolethal distending toxin from Shiga toxin-producing Escherichia coli O157 causes irreversible G2/M arrest, inhibition of proliferation, and death of human endothelial cells. Infect. Immun. 2005, 73, 552–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Lass, A.; Kujawa, M.; McConnell, E.; Paton, A.W.; Paton, J.C.; Wójcik, C. Decreased ER-associated degradation of alpha-TCR induced by Grp78 depletion with the SubAB cytotoxin. Int. J. Biochem. Cell Biol. 2008, 40, 2865–2879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Amaral, M.M.; Sacerdoti, F.; Jancic, C.; Repetto, H.A.; Paton, A.W.; Paton, J.C.; Ibarra, C. Action of Shiga toxin type-2 and subtilase cytotoxin on human microvascular endothelial cells. PLoS ONE 2013, 8, e70431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Kennedy, C.-A.; Fanning, S.; Karczmarczyk, M.; Byrne, B.; Monaghan, Á.; Bolton, D.; Sweeney, T. Characterizing the multidrug resistance of non-O157 Shiga toxin-producing Escherichia coli isolates from cattle farms and abattoirs. Microb. Drug Resist. 2017, 23, 781–790. [Google Scholar] [CrossRef] [PubMed]
  111. Valat, C.; Haenni, M.; Saras, E.; Auvray, F.; Forest, K.; Oswald, E.; Madec, J.-Y. CTX-M-15 Extended-spectrum β-lactamase in a Shiga toxin-producing Escherichia coli isolate of serotype O111:H8. Appl. Environ. Microbiol. 2012, 78, 1308–1309. [Google Scholar] [CrossRef] [Green Version]
  112. Arvand, M.; Bettge-Weller, G.; Fruth, A.; Uphoff, H.; Pfeifer, Y. Extended-spectrum beta-lactamase-producing Shiga toxin gene (stx1)-positive Escherichia coli O91:H14 carrying BlaCTX-M-15 on an IncI1-ST31 plasmid isolated from a human patient in Germany. Int. J. Med. Microbiol. 2015, 305, 404–407. [Google Scholar] [CrossRef] [PubMed]
  113. Day, M.; Doumith, M.; Jenkins, C.; Dallman, T.J.; Hopkins, K.L.; Elson, R.; Godbole, G.; Woodford, N. Antimicrobial resistance in Shiga toxin-producing Escherichia coli serogroups O157 and O26 Isolated from human cases of diarrhoeal disease in England, 2015. J. Antimicrob. Chemother. 2017, 72, 145–152. [Google Scholar] [CrossRef] [Green Version]
  114. World Health Organization (WHO) Critically Important Antimicrobials Human Medicine. Available online: https://apps.who.int/iris/bitstream/handle/10665/312266/9789241515528-eng.pdf?ua=1 (accessed on 1 April 2021).
  115. World Organization for Animal Health (OIE) OIE List of Antimicrobial Agents of Veterinary Importance. Available online: https://www.oie.int/fileadmin/Home/esp/Our_scientific_expertise/docs/pdf/AMR/E_OIE_Lista_antimicrobianos_Julio2019.pdf (accessed on 1 April 2021).
  116. Servicio Agrícola y Ganadero de Chile (SAG) Resolución N°1992. 5 Mayo 2006. Available online: https://www.bcn.cl/leychile (accessed on 1 April 2021).
  117. Servicio Agrícola y Ganadero de Chile (SAG) Resolución Exenta No:4854/2019, Modifica Resolución Exenta N° 6.801, de 2017. Available online: http://www.sag.cl/sites/default/files/resol_4.854-2019.pdf (accessed on 1 April 2021).
  118. Normativa y Avisos Legales de Uruguay (IMPO) Decreto N° 98/011 Prohibicion Del Uso de Antibioticos En La Alimentacion Para Animales Ovinos y Bovinos. Available online: https://www.impo.com.uy/bases/decretos/98-2011/1 (accessed on 1 April 2021).
Figure 1. Distribution of sequence types (STs) of Shiga toxin-producing Escherichia coli (STEC) strains circulating at the livestock-beef-environment-human interface, in South America. In Brazil, for livestock origin, ST25, ST642, ST1725, ST2522, and ST7100 were isolated from sheep feces, whereas ST223 was isolated from bovine, and ST2491 was isolated from pig feces [21,39,52,53,54,55].
Figure 1. Distribution of sequence types (STs) of Shiga toxin-producing Escherichia coli (STEC) strains circulating at the livestock-beef-environment-human interface, in South America. In Brazil, for livestock origin, ST25, ST642, ST1725, ST2522, and ST7100 were isolated from sheep feces, whereas ST223 was isolated from bovine, and ST2491 was isolated from pig feces [21,39,52,53,54,55].
Animals 11 01845 g001
Figure 2. Minimum spanning tree for 130 STEC strains isolated from cattle, beef, and humans in South America, built by the MSTree V2 tool and wgMLST scheme from Enterobase. Escherichia coli sequence types (ST), country, year, and source of origin are compared. Genome IDs in red letters correspond to strains sequenced in this study. Numbers in the branches show the differences of wgMLST alleles between strains. An interactive version of the tree can be found at https://itol.embl.de/tree/179113207189378131606509422.
Figure 2. Minimum spanning tree for 130 STEC strains isolated from cattle, beef, and humans in South America, built by the MSTree V2 tool and wgMLST scheme from Enterobase. Escherichia coli sequence types (ST), country, year, and source of origin are compared. Genome IDs in red letters correspond to strains sequenced in this study. Numbers in the branches show the differences of wgMLST alleles between strains. An interactive version of the tree can be found at https://itol.embl.de/tree/179113207189378131606509422.
Animals 11 01845 g002
Figure 3. SNP-based phylogenetic tree of the 21 STEC strains isolated from cattle samples collected in Chile (this study). The heatmap was constructed based on Clermont phylogroup, multilocus sequence typing (MLST), serotype, and presence/absence of Shiga-toxin subtypes and virulence, LAA-associated genes, and resistance genes. * Strain B-16 belongs to the novel ST-1661, a single locus variant (SLV) of ST33 at the fumC allele. ST11238 is an SLV of ST657 at the recA allele. SNP matrix is quoted in Table S3.
Figure 3. SNP-based phylogenetic tree of the 21 STEC strains isolated from cattle samples collected in Chile (this study). The heatmap was constructed based on Clermont phylogroup, multilocus sequence typing (MLST), serotype, and presence/absence of Shiga-toxin subtypes and virulence, LAA-associated genes, and resistance genes. * Strain B-16 belongs to the novel ST-1661, a single locus variant (SLV) of ST33 at the fumC allele. ST11238 is an SLV of ST657 at the recA allele. SNP matrix is quoted in Table S3.
Animals 11 01845 g003
Table 1. Logistic regression models for stx1 and stx1 + stx2 association with geographical location and isolation source of the 130 STEC whole genome sequencing (WGS) analyzed.
Table 1. Logistic regression models for stx1 and stx1 + stx2 association with geographical location and isolation source of the 130 STEC whole genome sequencing (WGS) analyzed.
stx195% CI
VariableCategoriesOdds Ratiop-ValueLowerUpper
Intercept 0.2380.0590.0541.057
Geographical locationArgentinaReference
Chile0.6570.6400.1133.832
Other3.9970.0600.94416.914
Isolation sourceBeefReference
Cattle3.8000.0171.26511.419
Human1.0690.9350.2145.347
stx1 + stx295% CI
VariableCategoriesOdds Ratiop-ValueLowerUpper
Intercept 0.1760.0460.0320.968
Geographical locationArgentinaReference
Chile0.6880.7140.0945.061
Others8.5410.0371.14263.859
Isolation sourceBeefReference
Cattle4.1250.0201.25513.557
Human0.2150.1360.0281.619
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Galarce, N.; Sánchez, F.; Escobar, B.; Lapierre, L.; Cornejo, J.; Alegría-Morán, R.; Neira, V.; Martínez, V.; Johnson, T.; Fuentes-Castillo, D.; et al. Genomic Epidemiology of Shiga Toxin-Producing Escherichia coli Isolated from the Livestock-Food-Human Interface in South America. Animals 2021, 11, 1845. https://doi.org/10.3390/ani11071845

AMA Style

Galarce N, Sánchez F, Escobar B, Lapierre L, Cornejo J, Alegría-Morán R, Neira V, Martínez V, Johnson T, Fuentes-Castillo D, et al. Genomic Epidemiology of Shiga Toxin-Producing Escherichia coli Isolated from the Livestock-Food-Human Interface in South America. Animals. 2021; 11(7):1845. https://doi.org/10.3390/ani11071845

Chicago/Turabian Style

Galarce, Nicolás, Fernando Sánchez, Beatriz Escobar, Lisette Lapierre, Javiera Cornejo, Raúl Alegría-Morán, Víctor Neira, Víctor Martínez, Timothy Johnson, Danny Fuentes-Castillo, and et al. 2021. "Genomic Epidemiology of Shiga Toxin-Producing Escherichia coli Isolated from the Livestock-Food-Human Interface in South America" Animals 11, no. 7: 1845. https://doi.org/10.3390/ani11071845

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop