Next Article in Journal
The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health
Next Article in Special Issue
In Vitro Microevolution and Co-Selection Assessment of Florfenicol Impact on Escherichia coli Resistance Development
Previous Article in Journal
Protective Effect of Procyanidin-Rich Grape Seed Extract against Gram-Negative Virulence Factors
Previous Article in Special Issue
Consumer Preferences and Attitudes towards Antibiotic Use in Food Animals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Resistance in Commensal Escherichia coli of the Porcine Gastrointestinal Tract

by
Lorcan O’Neill
1,2,*,
Edgar García Manzanilla
1,2,
Daniel Ekhlas
1,2,3 and
Finola C. Leonard
2
1
Pig Development Department, Teagasc, The Irish Food and Agriculture Authority, Moorepark, Fermoy, Co Cork P61 C996, Ireland
2
School of Veterinary Medicine, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
3
Food Safety Department, Teagasc Food Research Centre, Ashtown, Dublin D15 DY05, Ireland
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(11), 1616; https://doi.org/10.3390/antibiotics12111616
Submission received: 18 September 2023 / Revised: 7 November 2023 / Accepted: 9 November 2023 / Published: 11 November 2023

Abstract

:
Antimicrobial resistance (AMR) in Escherichia coli of animal origin presents a threat to human health. Although animals are not the primary source of human infections, humans may be exposed to AMR E. coli of animal origin and their AMR genes through the food chain, direct contact with animals, and via the environment. For this reason, AMR in E. coli from food producing animals is included in most national and international AMR monitoring programmes and is the subject of a large body of research. As pig farming is one of the largest livestock sectors and the one with the highest antimicrobial use, there is considerable interest in the epidemiology of AMR in E. coli of porcine origin. This literature review presents an overview and appraisal of current knowledge of AMR in commensal E. coli of the porcine gastrointestinal tract with a focus on its evolution during the pig lifecycle and the relationship with antimicrobial use. It also presents an overview of the epidemiology of resistance to extended spectrum cephalosporins, fluoroquinolones, and colistin in pig production. The review highlights the widespread nature of AMR in the porcine commensal E. coli population, especially to the most-used classes in pig farming and discusses the complex interplay between age and antimicrobial use during the pig lifecycle.

1. Introduction

Escherichia coli is the predominant aerobic microorganism in the microbiota of the vertebrate gastrointestinal tract [1,2]. Although primarily a commensal microorganism, several pathogenic strains are known to cause disease in both humans and animals [3]. Diarrhoeagenic strains of E. coli are an important cause of gastroenteritis in humans [4]. Some of these strains are zoonotic, with Shiga toxin-producing E. coli (STEC) representing the fourth most common bacterial food-borne infection in Europe, after campylobacteriosis, salmonellosis, and yersiniosis [5]. In humans, extraintestinal pathogenic E. coli (ExPEC) are the microorganisms most frequently implicated in urinary tract infections worldwide [6], and are the leading cause of bloodstream infections in Europe [7]. Antimicrobials are generally not indicated for the treatment of gastroenteritis caused by E. coli [8]. In contrast, they are essential in the treatment of ExPEC infections and, hence, antimicrobial resistance (AMR) in these bacteria is a major public health issue. Beta-lactams, cephalosporins (third generation and higher), fluoroquinolones, aminoglycosides, and carbapenems, all classified as critically important antimicrobials (CIA) by the World Health Organization (WHO) [9], represent the most important treatment options, and resistance to these classes in human isolates is monitored by the European Antimicrobial Resistance Surveillance Network (EARS-Net) [7]. This threat to public health is further highlighted by WHO’s inclusion of carbapenem and third-generation cephalosporin-resistant Enterobacteriaceae among its “Priority 1: critical” AMR pathogens for the development of new antibiotics [10].
Antimicrobial resistance in E. coli of animal origin poses a threat to human health in two ways. Firstly, similarities between certain ExPEC strains and avian pathogenic E. coli (APEC) strains in poultry have led some authors to suspect that some ExPEC infections may be zoonotic [11,12,13,14] and, in particular, associated with the consumption of chicken [15,16]. Secondly, as illustrated in Figure 1, animals represent a reservoir of AMR E. coli to which humans may be exposed via the food chain, direct contact with animals, or environmental contamination [17]. These AMR E. coli may transfer antimicrobial resistance genes (ARG) to bacteria of human importance by various means of horizontal gene transfer (HGT). Such HGT events may occur in the animal host, for example, between E. coli and Salmonella spp. [18,19], with subsequent zoonotic transmission; within the human host, after transient colonisation by E. coli of animal origin and subsequent HGT to human commensal or pathogenic bacteria [20]; or in the external environment. Thus, the commensal E. coli population is considered to be an indicator for AMR in the wider Gram-negative bacterial population. Pig farming is one of the largest livestock sectors worldwide [21] and is the highest consumer of veterinary antimicrobials [22]. Therefore, the transmission of AMR E. coli of porcine origin to humans via the food chain, occupational exposure, or environmental contamination poses a threat to public health. While recent whole genome sequencing (WGS) studies suggest that animals do not contribute significantly to the overall burdens of either ExPEC infections [23] or AMR E. coli in humans [24,25,26], the risks cannot be discounted entirely. The potential exists for new pathogenic strains to emerge, which would be especially concerning if accompanied by AMR. Moreover, occupational exposure to pigs has been associated with increased carriage of AMR E. coli on Canadian farms [27], while in the Netherlands, closely related extended spectrum beta lactamase (ESBL) producing strains of E. coli and/or ESBL genes were identified in farm and abattoir workers and in the pigs to which they were exposed [28,29,30]. Therefore, a thorough understanding of the dynamics of AMR E. coli in the pig lifecycle is required to help mitigate current and future threats to public health.
It should be noted that AMR in porcine bacteria is not limited to E. coli. Indeed, AMR is reported in a range of commensal and pathogenic bacteria, as can be seen in the reports of various national and international surveillance programmes [31,32,33], which include Salmonella spp., Enterococcus spp., Campylobacter jejuni, Staphylococcus aureus, Actinobacillus pleuropneumoniae, and Brachyspira hyodysenteriae, among others. Nor is AMR in porcine E. coli limited to commensal strains. Enterotoxigenic E. coli (ETEC) strains are implicated in neonatal and post-weaning diarrhoea in piglets, and treatment is frequently complicated by the presence of AMR [34,35]. However, this review focuses on AMR in commensal E. coli isolated from the gastrointestinal tract of healthy pigs. Published reviews on this topic include a systematic review analysing the relationship between AMR in E. coli and oral exposure to antimicrobials [36]; two recent systematic reviews examining resistance to extended spectrum cephalosporins, carbapenems, fluoroquinolones, and colistin [37,38]; and a review of AMR E. coli in China [39]. This literature review presents an overview and appraisal of current knowledge of AMR in commensal E. coli isolated from the porcine gastrointestinal tract in pig farming more generally, with a particular focus on its evolution during the pig’s lifecycle and the relationship with antimicrobial use (AMU). A brief review of the epidemiology of resistance to extended spectrum cephalosporins, fluoroquinolones, and colistin in pig production is also presented.
Before presenting the review, it is important to consider the criteria used to interpret the antimicrobial susceptibility tests (AST), which determine the presence or absence of AMR. Different interpretive criteria can hamper direct comparison between studies, especially when quantitative data, i.e., minimum inhibitory concentrations (MIC) or zone diameters, are not available. Some of the studies presented here used clinical breakpoints (CBP) as their interpretive criteria. These are determined using MIC distribution, pharmacokinetic/pharmacodynamic and clinical outcome data, and thus describe ‘clinical resistance’ [40]. Other studies used epidemiologic cut off values (ECOFF) which divide the bacterial population according to the presence, i.e., non-wild type (nWT), or absence, i.e., wild type (WT), of acquired resistance mechanisms. These are determined using MIC or zone diameter data only and thus describe ‘microbiological resistance’ [41]. It is important to distinguish between microbiological and clinical resistance because the presence of an acquired resistance mechanism (whether determined phenotypically or genotypically) is not always associated with clinical resistance and, as such, ECOFFs should not be used as a basis for clinical decisions [41]. Notwithstanding the distinction between clinical and microbiological resistance, many authors use the terms ‘susceptible’ and ‘resistant’ to describe WT and nWT isolates, respectively (with or without explaining the distinction) and, although not strictly correct, do so in the interest of readability. Clinical breakpoints and ECOFFs can differ depending on whether European Committee on Antimicrobial Susceptibility Testing (EUCAST), Clinical and Laboratories Standards Institute (CLSI), or national guidelines (CBPs only) are used [40,42,43,44]. Furthermore, different CBPs may be defined for humans and animals (some studies used veterinary breakpoints) and both CBPs and ECOFFs may change over time (meaning that older studies may not be directly comparable to more recent ones). Finally, some studies may use other criteria, usually for the purposes of screening for specific resistance mechanisms, which are not always analogous to the relevant ECOFF or CBP (e.g., specific monitoring of ESBL/AmpC producing E. coli in food animals in Europe [33,45]). The various interpretive criteria discussed highlight a limitation in the field of clinical microbiology, generally, and in this review, specifically, regarding the comparison of results between studies. However, it would be impossible to discuss all of the disparities between the different studies during the narrative without hampering readability. Therefore, for the purposes of this review, the terms susceptible and resistant refer to the interpretive criteria used by the authors of the individual studies, although the reader should keep in mind the nuances of AST and its interpretation as they proceed through the text.

2. Antimicrobial Resistance in Escherichia coli of Porcine Origin: Surveillance Programmes

Antimicrobial resistance in E. coli of porcine origin is not a recent phenomenon. In 1957, Smith and Crabb reported that the continuous inclusion of tetracyclines in feed selected for tetracycline resistance in the commensal E. coli population in pigs [46], and a further longitudinal study conducted in the years after a ban on the use of medically important antimicrobials as feed additives/growth promoters in the United Kingdom (UK) showed that relatively high levels of tetracycline, streptomycin, and sulphonamide resistance had persisted and that trimethoprim resistance had emerged by the end of the 1970s [47]. Similarly, in Denmark, Albaek et al. (1991) showed that, despite similar restrictions on the use of growth promoters, the levels of resistance to tetracycline, streptomycin, and sulphonamide were significantly higher in 1988 than those reported in two earlier studies conducted in the 1970s [48]. Moreover, the prevalence of resistance to ampicillin, neomycin, and chloramphenicol, which were rare in the earliest of the comparison studies (3%, 0% and 3%, respectively), had also increased (84%, 47% and 30%, respectively) by 1988 [48]. More recently, commensal E. coli from food animals, including pigs, have been included with pathogenic bacteria in the AMR monitoring programmes of several countries, in line with recommendations by the WHO Advisory Group on Integrated Surveillance of Antimicrobial Resistance (AGISAR) [49]. Examples of these programmes outside of Europe include the National Antimicrobial Resistance Monitoring System (NARMS) in the USA [50], the Canadian Integrated Program for Antimicrobial Resistance Surveillance (CIPARS) [51], and the Japanese Veterinary Antimicrobial Resistance Monitoring System (JVARM) [52]. In Europe, the European Food Safety Authority (EFSA) and the European Centre for Disease Prevention Control (ECDC) oversee a programme that mandates European Union (EU) member states to monitor AMR in commensal E. coli from slaughter pigs biennially [33]. Some non-EU states also participate (e.g., European Economic Area (EEA) members, Switzerland) and, while the UK no longer participates since leaving the EU, it continues to publish its United Kingdom Veterinary Antibiotic Resistance and Sales Surveillance (UK-VARSS) reports using the same methodology [31]. Another pan-European programme, the European Antimicrobial Susceptibility Surveillance in Animals (EASSA), is operated by the pharmaceutical industry [53]. Several European states also operate their own monitoring programmes, for example, the Danish Integrated Antimicrobial Resistance Monitoring and Research Programme (DANMAP) in Denmark [54] and the Monitoring of Antimicrobial Resistance and Antibiotic Usage in Animals in the Netherlands (MARAN) programme in the Netherlands [55]. As these monitoring programmes mature, more detailed analyses, e.g., of multiyear trends, will become available, as is the case for Portugal, Denmark, and the USA [56,57,58]. Schrijver et al. (2018) reviewed the monitoring programmes in operation in Europe up until 2016 and found marked heterogeneity in sampling and laboratory methodology, as well as in the availability of results (i.e., language and frequency of publication) [59]. However, the EFSA and ECDC programme, with harmonised protocols and transparent reporting, allows for a comparison of AMR in the participating European countries. In contrast, monitoring programmes in lower- and middle-income countries (LMIC), including China, the largest pig producing country globally, are largely absent or at least not publicly available in the English language [60,61,62].

3. Resistance in Escherichia coli Isolated from Finisher Pigs: Data from Surveillance Programmes and Published Studies

The handling and consumption of pork is expected to represent the highest risk of human exposure to AMR E. coli of porcine origin in the general population. Therefore, national monitoring programmes and the majority of published studies sample pigs at or close to slaughter. Table 1 presents a summary of the most recent data from the EFSA and ECDC, UK-VARSS, NARMS, CIPARS, and JVARM programmes. These data show that resistance to tetracyclines, sulphonamides, trimethoprim, aminopenicillins, amphenicols, and aminoglycosides is common, reflecting the widespread use of these classes in pig production. Resistance to the highest priority critically important antimicrobials (HP CIA) [9] is generally lower, although high levels of fluoroquinolone resistance are observed in some individual European nations. It should be noted that, while streptomycin, an aminoglycoside, is not included in the EFSA and ECDC testing panel, relatively high levels of resistance are expected, in line with cross-sectional European studies [63,64,65,66,67]. High levels of streptomycin resistance, not evaluated in the most recent NARMS data but included previously [68], are also reported in the USA [58]. Table 2 summarises the results from a selection of studies that investigated AMR in commensal E. coli isolated from healthy finisher pigs on farm or at slaughter. Not intended to present an exhaustive list of all such studies carried out, comparisons between the studies included in this table should be made with caution. As discussed in the introduction, the criteria used to interpret the AST results can hamper direct comparison between studies. Differences in study design, for example sample size, the age of the animals at sampling, or the laboratory methods employed, may also impact the results obtained. Nevertheless, the results from the European and North American studies in Table 2 are broadly in line with those from their respective regional monitoring programmes, which, along with the results from other regions, confirm high prevalence of resistance to the heavily used antimicrobial classes in most settings, with some exceptions. In Europe, northern countries such as Sweden and Norway, with long histories of good antimicrobial stewardship, generally have lower AMR prevalence than southern European countries [33]. In Africa, AMR was generally lower in the less intensive production systems studied in Nigeria, Uganda, and Rwanda [69,70,71] than the intensive systems in Tanzania [72,73]. On the other hand, studies from LMICs in Asia suggest that AMR in intensive pig production is higher than in developed countries, particularly for HP CIAs. In China, the high rates of resistance to tetracyclines, sulfonamides, beta-lactams, amphenicols, and fluoroquinolones illustrated in Table 2 [74,75] are consistent with other studies in which the age and/or health status of the sampled pigs were uncertain [76,77,78,79,80,81] and with the findings of a recent meta-analysis [39]. They are also consistent with the estimated resistance rates determined in a pooled analysis of point prevalence studies that investigated AMR in E. coli isolated from pig farms, slaughterhouses, and food in China between 2000 and 2019 [62]. Interestingly, there is disagreement between some of the studies conducted in China concerning resistance to third-generation cephalosporins and to colistin. For example, colistin resistance was high in some studies, for example 46.3% [76] and 59.7% [82], but low or absent in others [74,77,80,81]. Such discrepancies could be due to differences in geographical area in such a large country, study design, or laboratory methods, and further highlight the benefits of systematic surveillance. In Southeast Asia, two studies in Thailand that investigated AMR in finisher pigs on groups of farms with different levels of AMU also reported high rates of resistance to most of the antimicrobials tested, especially on farms with routine prophylactic AMU, where almost all isolates were resistant to tetracycline, ampicillin, trimethoprim/sulfamethoxazole, and chloramphenicol [83,84]. Furthermore, rates of resistance to third-generation cephalosporins and fluoroquinolones in those studies were well in excess of those reported in developed countries. Similar results were reported in studies conducted at slaughterhouses in Vietnam, Cambodia, and Thailand [85,86], highlighting concerns that LMICs, especially in Asia, represent ‘hotspots’ of AMR [37,38,61].

4. Resistance in Escherichia coli Isolated from Pigs in Age Groups Other Than Finisher

Much of the work investigating AMR in commensal E. coli in younger pigs involved experimental trials. These often involved using specific antimicrobial treatments [103] or were conducted on experimental farms or over limited timeframes [104], meaning that their findings may not be directly applicable to conditions in the field. Compared with the number of field studies investigating AMR in finisher pigs, relatively few have investigated AMR in commensal E. coli originating from other age groups. Many of these studies reported higher levels of AMR in younger animals, which is a phenomenon noted in various other species, including humans [105]. However, this varied somewhat between studies as temporal trends may differ depending on the antimicrobial and/or AMU patterns studied. Younger pigs, especially after weaning, are more likely to receive antimicrobials [106,107] which undoubtedly affects the prevalence of resistance. Studies in Ireland, Canada, USA, and Spain all showed higher rates of resistance in isolates from weaner pigs compared with finisher pigs for most, if not all, of the antimicrobials studied. [65,96,108,109]. Similarly, Pissetti et al. (2021) reported increased odds of a multidrug resistant (MDR) phenotype in isolates from nursery pigs compared with finisher pigs on Brazilian farms [110]. While the peak in AMR in E. coli during the weaner stage reported in these studies coincides with high AMU in this age group, the findings of other studies suggest that AMU alone may not explain this phenomenon. On a US research farm not exposed to antimicrobials for over five years, resistance to tetracycline, sulfisoxazole, and streptomycin was higher in isolates from weaner pigs compared with most of the older age groups [111]. More recently, Yun et al. (2021) reported that the prevalence of MDR on 10 Finnish farms was higher at 5 weeks of age compared with 22 weeks, regardless of whether they received antimicrobials during their lifetime or not [112]. A longitudinal study conducted on 29 farms in Germany tracked resistance to ampicillin, tetracycline, colistin, and azithromycin in pigs treated or not treated with the respective antimicrobial class [113]. Resistance to ampicillin and tetracycline in untreated pigs, as well as to azithromycin regardless of treatment status, peaked during the weaner stage. In contrast, the lowest levels of resistance were observed in finisher pigs with the exception of tetracycline resistance in tetracycline-treated pigs. In that study, the higher tetracycline resistance in treated finisher pigs was likely associated with tetracycline use in the later production stages, whereas the other antimicrobial classes were only used in younger pigs [113]. A cross-sectional study in the USA compared AMR in antibiotic free (ABF) and conventional herds (n = 3 and n = 4, respectively) [109] and found that the minimum inhibitory concentrations (MIC) of ampicillin, gentamicin, and sulfamethazine were highest in the two youngest age groups (pigs weighing 4.5 kg and 23 kg), but only on conventional farms. This was not the case for oxytetracycline, where resistance on the ABF farms was highest in the younger pigs, despite the absence of AMU, but highest in finisher pigs from conventional farms who all used antimicrobials (including tetracyclines) during the finisher stage [114]. In contrast, Græsbøll et al. (2017) found that tetracycline resistance was lower just before slaughter than at earlier time points, although the differences were not statistically significant, and that a significant post weaning peak in resistance was only observed in groups treated with oxytetracycline [115]. These studies provide evidence of the influence of age on AMR, which appears to be independent of AMU. Nevertheless, given the relationship between age and AMU, and the age-related dynamics of the E. coli population in the porcine intestinal tract [116,117,118], it is difficult to separate both factors. It is reasonable to attribute this age-related effect to an evolutionary response in the bacterial population, whereby E. coli in weaned pigs are adapted to a post weaning intestinal environment which is frequently exposed to antimicrobials. Extended spectrum cephalosporins and fluoroquinolones are notable exceptions to the post weaning peak in resistance, with the higher prevalence observed in piglets [65,119,120] likely influenced by higher exposure to these classes in this age group [107]. Resistance to these classes is discussed separately below.
Relatively few studies have investigated AMR in E. coli from sows. Sows represent an important reservoir of AMR on the farm and, although the sow and piglet E. coli populations are different [116], associations between resistance in E. coli from sows and piglets have been demonstrated in several studies [113,121,122,123,124]. Burow et al. (2019) reported similar levels of resistance in sows and finishers in their study and significant associations between resistance to ampicillin and azithromycin in sows and piglets [113]. In Ireland, the prevalence of AMR in E. coli originating from sows was similar to piglets for most of the antimicrobials studied, but higher than in finishers [65]. In contrast, Mathew et al. (2001) reported lower AMR in sows compared with finishers, especially in the ABF herds [114]. Sows on Swiss farms had a lower prevalence of resistance compared with weaners for most of the antimicrobials studied, although not for ciprofloxacin [125]. Two cross-sectional studies in north-eastern Thailand that sampled only sows showed high rates of resistance [126,127] consistent with other studies that sampled finisher pigs in the region [80,84,128]. The NARMS programme includes sampling of sows at slaughter [68] and a recent analysis of the data between 2013 and 2019 showed a generally higher prevalence of AMR in E. coli isolated from finisher pigs compared with sows [129]. Such findings contrast with the previously mentioned associations between AMR in sows and their offspring. This apparent contradiction may be influenced by the timing of sampling, i.e., whether sows are lactating or gestating, but studies investigating whether parturition and/or lactation and their associated stresses have an effect on AMR are lacking.

5. Mechanisms of Antimicrobial Resistance in Escherichia coli

An understanding of the resistance mechanisms employed by E. coli is useful in explaining the underlying epidemiology of AMR. Overall, accurate estimates of resistance gene prevalence in the porcine E. coli population are difficult to infer from the literature because genotypic studies in the field are carried out less frequently than phenotypic studies, and the methodology may differ in terms of the type of animal sampled and the profile of isolates chosen for evaluation. Furthermore, studies that use PCR methods rely on which gene(s) are chosen for study and thus depend on prior knowledge of the prevailing genotypes. Such prior knowledge is not required for whole genome sequencing (WGS), which allows for an accurate characterisation of the AMR genotype. While studies using WGS to investigate the generic E coli commensal population in pigs are still relatively rare, the NARMS programme has performed WGS on a subset of its isolates [68] since 2017, and studies from Europe, Spain and the UK on finisher pigs [130,131,132,133] and from Australia on weaner pigs [134,135] have been published recently. Another study performed a retrospective in silico analysis of E. coli WGS data from livestock, including pigs, retrieved from three publicly available genome databases [136]. The latter study reported increasing trends in the prevalence of some ARGs, such as tet(A), blaTEM-1b, blaCMY-2, and floR, over the study period from 1980–2018, although the lack of farm metadata and information on aspects such as sample site or the age of the animal at sampling precludes any farm level analysis of the molecular epidemiology of these resistance mechanisms [136]. The main AMR mechanisms utilised by E. coli of animal origin were reviewed by Poirel et al. (2018) [137]. In pigs, tetracycline resistance is most often conferred by the efflux genes tet(A) and/or tet(B) [68,130,131,132,133,134,135,136], although others such as tet(M), a ribosomal protective gene, have been reported [68,81,132,133,138]. Interestingly, Pires et al. (2021) noted the displacement of tet(B) by tet(A) between 1980 and 2018 in the collection of isolates studied [136], a finding that highlights the dynamic nature of ARG epidemiology. The association of the narrow spectrum beta-lactamase blaTEM-1 gene (particularly the 1b variant) with ampicillin resistance is an almost universal finding in the studies investigating it [104,130,131,132,133,136,139]. Genes conferring resistance to sulphonamides and trimethoprim, which interfere with bacterial folate metabolism, are also widespread in the porcine E. coli population. The prevalence of sul1, sul2, and sul3 varies between studies; sul2 was the most common gene in a UK cross sectional study [132] and in the NARMS data [68], sul3 was the predominant gene in a Thai study [128], whereas both sul2 and sul3 were common on Spanish farms [133]. Streptomycin resistance is associated with the strA/strB gene pair and with the aadA genes (which also confer resistance to spectinomycin); both groups are commonly found on pig farms [130,132,133]. Amphenicol resistance is mainly conferred by the catA and cmlA genes [140], which persist in the E. coli population, despite the fact that chloramphenicol is no longer used in animals in most countries. The floR gene is important in the veterinary context as it confers resistance to both florfenicol, which is used in pig farming, and chloramphenicol. Florfenicol resistance is not always reported for studies of commensal isolates, but appears widespread in China [39,141]. In contrast, floR was found in 5.5%, 2.9%, and 11.7% of isolates in the UK, USA, and Spain respectively [68,132,133]. The genes conferring resistance to tetracyclines, sulphonamides, trimethoprim, aminoglycosides, and amphenicols represent the most widely distributed ARGs in the E. coli population and are frequently co-located. Such co-location explains the phenomenon of multidrug resistance. Indeed, multidrug resistance is a notable feature of many of the studies included in this review and, as an illustrative example, 30.3% of all E. coli isolated from pigs at slaughter submitted to the EFSA and ECDC monitoring programme in 2021 were MDR, 44.2% of which were resistant to tetracycline, ampicillin, sulfamethoxazole, and trimethoprim [33]. The relevant ARGs are usually located on mobile genetic elements (MGE) and, in fact, are often co-located on the same MGE. Integrons, especially class 1, have an important role in the epidemiology of MDR [142] as they can capture, express, and exchange ARG cassettes [143] and are frequently located on transposons or plasmids that can facilitate HGT [144]. The most common integron-associated gene cassettes include variants of aadA and dfrA, but others such cmlA are prevalent in pigs [135,145]. Furthermore, the sul1 gene is part of the conserved 3′ region of the classical class 1 integron structure and sul3 is also associated with integrons [134,135,146]. This means that isolates harbouring integrons are commonly resistant to sulphonamides, trimethoprim, aminoglycosides, and/or amphenicols. Several studies, especially in Asia, have reported a high prevalence of class 1 integrons of up to 75% in E. coli isolates of porcine origin [75,101,105,147,148]. Moreover, although tet and blaTEM are not found on integrons, they are frequently found in integron positive isolates [147].
Despite their abundance (and indeed, because of it), the resistance mechanisms discussed so far are less important in terms of public health, since the associated drugs are no longer routinely used in the treatment of Enterobacteriaceae infection of humans. On the other hand, antimicrobials such as third-generation (and higher) cephalosporins, fluoroquinolones, gentamicin, and carbapenems are essential in the treatment of Gram-negative infections and, thus, their respective resistance mechanisms are topics of considerable interest. Gentamicin is a medically important aminoglycoside antibiotic and several genes conferring resistance in E. coli have been reported [137]. The aac(3)-IVa gene is especially important in the veterinary context as it confers resistance to the veterinary drug apramycin and to gentamicin [137], and its occurrence in human clinical isolates illustrates a rather concrete example of the interface between AMR in animals and humans [149]. The aac(3)-IVa gene was the most prevalent gentamicin resistance gene detected in a WGS study carried out on UK finisher herds [132], in two studies in Australia [134,135], and in two older studies in Denmark [150] and Korea [151]. In contrast, the aadB gene was common on Thai pig farms [83,128] (the authors did not investigate aac(3)-IV), while the NARMS WGS data suggests that aac(3)-IId is more prevalent in finisher pigs in the USA [68]. Interestingly, AbuOun et al. (2020) also detected aac(3)-IId, but only in fluoroquinolone-resistant isolates recovered from selective media [132]. Tigecycline is a synthetic derivative of tetracycline, developed to treat MDR infections [152]. Resistance, associated with the tet(X) ARG, was first reported in isolates of animal origin in China [153], and has been detected in E. coli isolates of porcine origin in a number of Chinese studies [80,81] and in five European countries participating in the EFSA and ECDC monitoring programme in 2021 [33]. Carbapenems are of the utmost importance in human medicine as antimicrobial agents of last resort. While still relatively uncommon, resistance genes such as blaVIM and blaNDM are reported in E. coli of porcine origin [33,37,81]. Resistance to the extended spectrum cephalosporins, fluoroquinolones, and polymyxins are discussed separately below.

6. Relationship between AMU in Pig Farming and AMR in Escherichia coli

6.1. Ecological Associations between AMU and AMR in Escherichia coli

Antimicrobial use is generally accepted to be the main driver of AMR and, therefore, the relationship between AMU and AMR in animals is a topic of considerable interest. Ecological studies have identified associations between AMU at a national level and AMR in E. coli recovered from national monitoring programmes. Chantziaras et al. (2014) found that the use of specific antimicrobial classes at a national level in seven European countries was correlated with the level of resistance in commensal E. coli in cattle, pigs, and poultry [154]. At an individual country level, a trend analysis on data from Belgium between 2011 and 2015 found significant associations between resistance and use of the corresponding class for 10 out the 11 classes studied, while resistance was associated with total AMU for all classes [155]. Both of these studies used species aggregated data and so direct inference for the pig-related data is not possible. However, a Japanese study reported significant correlations between the prevalence of resistance to specific antimicrobial classes in the pig sector and the consumption of the corresponding classes [156], and in Denmark, gentamicin resistance was associated with the consumption of apramycin [152]. More recently, the ‘joint inter-agency reports on integrated analysis of antimicrobial agent consumption and occurrence of antimicrobial resistance in bacteria from humans and food-producing animals in EU/EEA’ (JIACRA) used AMU surveillance data from the European Surveillance of Veterinary Antimicrobial Consumption (ESVAC) project along with AMR data from the EFSA and ECDC monitoring programme to explore the relationship between AMU in livestock and AMR in E. coli in Europe [157]. These analyses found significant associations between resistance to fluoroquinolones, colistin, ampicillin, and tetracycline, in indicator E. coli, and the use of the corresponding antimicrobial classes at a country level for both the pig specific and the aggregated species datasets [157]. A pan-European study carried out by the EFFORT consortium that sampled 180 farms in nine countries also found significant associations between AMR in E. coli and the average treatment incidence at a country level [158]. These associations included cephalosporin use with ampicillin resistance, fluoroquinolone use with ciprofloxacin and nalidixic acid resistance, amphenicol use with chloramphenicol resistance, and lincosamide use with azithromycin resistance [158]. Dorado-Garcia et al. (2016) used species-specific data for a similar analysis in the Netherlands and found that resistance in porcine isolates to a particular antimicrobial was associated with total use rather than use of the corresponding class [159]. Taken together, these studies demonstrate a relationship between AMR in E. coli and background AMU at country level. Moreover, they demonstrate that reductions in AMU can lead to reductions in AMR. The Dutch and Belgian studies mentioned previously were conducted during a period of declining AMU in livestock [155,159], and there are similar examples for colistin resistance in China where resistance in E. coli and the prevalence of the associated mcr-1 gene in humans and animals (including pigs) reduced substantially following a ban on the use of colistin as an antimicrobial growth promoter (AGP) [160,161].

6.2. Intervention Studies Investigating Relationship between AMU and AMR

Studies investigating the relationship between AMU and AMR can be divided into two categories: intervention studies where the effect of administering an antimicrobial on AMR is investigated, and observational studies where a cohort (or cohorts) of farms are sampled and the results are analysed in conjunction with farm-level AMU data. The former category of study usually takes place on a research farm or on a limited number (often single) of commercial farms, which may allow for a controlled environment, although it may not reflect conditions in the field. Langlois et al. (1978) examined the effect of five in-feed antimicrobial protocols (untreated, bacitracin, virginiamycin, tylosin, and chlortetracycline) on resistance to chlortetracycline (CTC) and found that resistance was lowest in the untreated groups and highest in the groups treated with CTC [162]. In another study, Langlois et al. (1984) found that the response to antimicrobial treatment was affected by the farm’s antimicrobial exposure history [163]. That study evaluated the effect of sub therapeutic and therapeutic doses of CTC on tetracycline resistance in two herds of pigs, one of which was from an ABF farm, and found a greater increase in resistance in the treated groups from the ABF herd compared with the treated groups from the herd with antibiotic exposure [163]. Other studies have demonstrated similar results along with an increase in resistance to unrelated antimicrobials [103,115,164,165]. Delsol et al. (2003) found that tetracycline resistance reduced after treatment was withdrawn, but remained above pre-treatment levels for at least two weeks [164]. Græsbøll et al. (2017) had similar findings, but reported that resistance returned to pre-treatment levels prior to slaughter [115]. A US study investigating resistance to apramycin found that apramycin resistance persisted for longer after apramycin treatment if the pigs had previously been treated with oxytetracycline [166]. Levels of AMR and its persistence were also affected by antimicrobial exposure in the sows [121] and other environmental factors such as temperature, stocking density, and mixing [166,167,168]. These findings are relevant to field studies as there may be a wide variation between farms and, indeed, many of these factors may not be recorded or measurable. Some studies investigated the effect of different treatment regimens in terms of dose and or route of administration. This is of interest because identifying a mode of treatment associated with a lower risk of antimicrobial resistance would be beneficial. Overall, however, the evidence is limited and somewhat contradictory [36]. Increased doses of apramycin were associated with higher aminoglycoside resistance in one study [169]. On the other hand, there was no difference in the response to treatment with different doses of oral or injectable oxytetracycline on five Danish farms [115], which agreed with the conclusions of another Danish study [170]. Similarly, ampicillin resistance in E. coli was similar in groups of pigs treated with oral or injectable ampicillin [104]. Interestingly, while the prevalence of phenotypic resistance within Enterobacteriaceae did not vary between treatment groups in the latter study, there were higher Enterobacteriaceae plate counts and higher gene copy numbers of blaTEM detected by qPCR in the faeces of the oral treatment groups [104]. This suggests that mode of treatment affected the Enterobacteriaceae and other members of the microbiota differently, and highlights how the quantification of AMR may be subject to different interpretations depending on which methods are used and on the subject population under study. It is also worth noting that this experiment found a rise in multidrug resistance in the E. coli population in response to treatment, and that this change was associated with a shift in the phylogenetic profile of the E. coli population to phylogroups possessing ampicillin and MDR phenotypes [171]. Interestingly, some studies observed increases in AMR in untreated animals kept in the same pens or rooms as treated animals [115,120,172,173,174]. In one of these studies, fluoroquinolone-resistant E. coli strains similar to those found in the fluoroquinolone-treated group were detected in the untreated control group, even though both groups were housed in separate pens [174]. Such findings demonstrate that AMR is influenced by AMU in the community as well as in the individual, and further demonstrate the complexity of AMR epidemiology. Taken together, the intervention studies discussed demonstrate that, in general, antimicrobial treatment causes a rise in AMR in the E. coli population and is followed by a decline to pre-treatment levels (in the studies in which this was measured). While these studies provide valuable information on the dynamics of AMR, the findings are not always applicable to the situation in the field, especially as they are usually carried out on a single herd on research farms.

6.3. Observational Studies Investigating Relationship between AMU and AMR

Observational studies allow for investigation of the relationship between AMU and AMR at farm level. Ideally, the AMU data used in such studies are as detailed and complete as possible. In practice, such data are not always available and thus categorisation of AMU must be used (e.g., use or not of a particular antimicrobial, high AMU vs. low AMU). Gellin et al. (1989) compared three university farm herds, one that routinely used AGPs, one that only used antimicrobials therapeutically when required, and, lastly, one that did not use any antimicrobials [175]. In almost all cases, resistance to each of the antimicrobials studied was highest on the farm using AGPs and higher on the therapeutic-use farm than the ABF farm [176]. A similar cross-sectional study carried out on seven farrow-to-finish farms, including three ABF farms and four conventional farms in the USA, had similar results [114]. Mathew et al. (1998) also found a higher resistance prevalence on high AMU farms compared with low AMU farms in a longitudinal study conducted from birth to nine weeks of age [176]. Bunner et al. (2007) sampled finisher pigs on 35 ABF and 60 conventional farms in the USA, and reported that the odds of resistance to all antimicrobials were higher on conventional farms, although resistance to quinolones and third generation cephalosporins was absent in both groups [97]. Resistance was also lower on organic farms from four European countries compared with their conventional counterparts [66] and on free range Iberian pig farms compared with conventional farms in Spain [177]. These studies provide evidence for a relationship between AMU and AMR, at least when comparing no or very limited use to higher AMU conventional farms. However, in Southeast Asia, the situation is not as clear cut. In one study in Thailand, resistance to sulfamethoxazole, gentamicin, and chloramphenicol was higher on medium-scale farms (>100 sows) compared with small-scale farms (<100 sows), but resistance to tetracycline was higher on the small-scale farms [127]. A companion study that used the same farms and samples as the previously mention study, but with different laboratory methods, found a higher prevalence of colistin resistance on the small-scale farms [178]. In another, unrelated, Thai study, resistance on farms practising prophylactic AMU was higher than on farms practising only therapeutic use or those with no AMU for most of the antimicrobials investigated, but not for tetracycline or ampicillin [83]. In that study, there were no differences between the therapeutic use farms and ABF farms, and the prevalence of resistance in both groups was, in most cases, higher than the equivalent prevalence on European or North American conventional farms. In fact, the ABF farms were all rural, small-scale farms with no apparent access to veterinary care of any kind [83]. In a similar, but larger-scale study in Thailand, resistance to cephalosporins, azithromycin, and colistin was higher in the prophylactic AMU group compared with those with lower AMU, but the prevalence of resistance to most of the other antimicrobials in all three groups was similar [84]. The high rates of AMR in ABF production systems in these studies could reflect very high background levels of resistance in the region or perhaps reflect unreported or unknown AMU in these herds.
Studies that report the use or not of particular antimicrobial agents or the amounts used allow for a more detailed analysis of the relationship between AMU and AMR. The prevalence of resistance to tetracycline in E. coli isolated from pigs of different age groups on Belgian farms was associated with the use of tetracyclines, as well as with the treatment incidence of potentiated sulphonamides [179], which demonstrates both direct and co-selection of resistance associated with AMU. Vieira et al. (2009) also reported a positive association between tetracycline use and tetracycline resistance in Danish pigs at slaughter [180]. Notably, there was a significant association between resistance and the length of time between treatment and slaughter [180], meaning that the closer the last tetracycline treatment was to slaughter, the higher the probability of resistance. Several more studies have reported associations between the use of specific antimicrobials and resistance to their respective classes, albeit not in all instances [65,181,182,183,184,185,186]. These studies also demonstrated associations between the use of specific drugs and resistance to unrelated antimicrobial classes. The use of in-feed antimicrobials in at least one diet was associated with resistance to six out of seven antimicrobials tested on 34 farms in Ontario [181]. The exception was gentamicin resistance, which was only associated with injectable gentamicin use. In four of these models, the association was not antimicrobial specific, meaning resistance increased regardless of which antimicrobial was used, and medicating the starter diet in early weaning was associated with resistance in the finisher stage [181]. The use of medicated feed in grower or finisher diets was also associated with AMR, including MDR, in isolates from finisher pigs in a study in Alberta for four of the investigated models [185]. Similar to the study of Dunlop et al. (1998) [181], there were antimicrobial-specific and non-specific associations with resistance, as well as examples of co-selection [185]. Interestingly, some studies demonstrated associations between macrolide use and unrelated antimicrobial classes, even though E. coli is considered intrinsically resistant to macrolides [65,182,185,187]. For example, macrolide use was associated with chloramphenicol resistance on Irish farms [65]. Overall, these studies provide evidence for a complex relationship between AMU and AMR in E. coli. Resistance is influenced by AMU both recently and earlier in the lifecycle, as well as by historical usage and background resistance. In many cases, the relationship between AMU and AMR is not clear cut, with some conflicting findings between studies; however, co-selection has an extremely important role. The persistence of chloramphenicol resistance despite a lack of use demonstrates how resistance genes can be maintained in the population due to their association with other resistance genes. An apparent relationship between macrolide use and resistance to other classes further illustrates this complex relationship. The majority of these studies are cross-sectional and, as discussed before, mostly sample pigs at or close to slaughter. A notable exception is the study by Burow et al. (2019), who followed pigs from birth to slaughter on 29 German pig farms [113]. In that study, E. coli isolates from pigs treated with penicillins, tetracyclines, polymyxins, or macrolides were more likely to be resistant to ampicillin, tetracycline, colistin, or azithromycin, respectively, than isolates from untreated pigs, but not at all timepoints [113]. In fact, only tetracycline had a significant difference between treated and untreated pigs at the last time point just before slaughter. This shows that studies conducted only in older pigs may not truly capture the relationship between AMU and AMR throughout the whole pig lifecycle.

7. Resistance to Extended Spectrum Cephalosporins, Fluoroquinolones, and Polymyxins

Extended spectrum cephalosporins (ESC), fluoroquinolones, and polymyxins (i.e., colistin) are classed as HP CIA by WHO (along with glycopeptides and macrolides) and as ‘Category B’ antimicrobials by the European Medicines Agency (EMA) [9,188]. Although not as heavily used as other antimicrobials and nowadays subject to restrictions in certain settings [189], they are important veterinary antimicrobials, and, in particular, fluoroquinolones and colistin have indications for the treatment of E. coli infections in animals. Resistance to these classes in commensal E. coli in livestock is especially important because of the importance of these drugs in treating Gram-negative infections in humans and the fact that resistance is well established in both human and animal populations. Isolates resistant to one of these antimicrobials are usually resistant to several other classes of antimicrobial, as illustrated in a study on Belgian poultry and pig farms, where over 90% of ciprofloxacin or cefotaxime-resistant isolates were MDR [190], and in two recent studies that reviewed and conducted meta-analyses of resistance to ESCs, fluoroquinolones, and colistin [37,38].
The main mechanisms of ESC resistance are extended spectrum beta lactamases (ESBL) and AmpC beta lactamases, which are, in most cases, associated with MGEs and plasmids [191,192,193]. In humans, ESBLs are the most prevalent and, of these, the CTX-M family is the most important, with the CTX-M-15 and CTX-M-14 variants being the most prominent globally [194,195,196]. Several studies have investigated the prevalence of ESBL/AmpC-producing E. coli in pigs, alone or in conjunction with other food producing species. In contrast with humans, there are more distinct geographical patterns regarding the distribution of ESBL/AmpC genes [37]. In East Asia, blaCTX-M-14, blaCTX-M-55 and blaCTX-M-65 predominate [197,198,199,200,201,202,203,204,205,206]. In North America, ESC resistance is usually conferred by the plasmid-mediated AmpC blaCMY-2 gene, as ESBLs are relatively rare in food-producing animals [37,68,207,208,209]. In Europe, data from the EFSA and ECDC specific monitoring of ESBL/AmpC producing E. coli show that the ESBL phenotype is more prevalent than the AmpC phenotype, although this varies between countries [33]. This component of the EFSA and ECDC monitoring programme aims to estimate the prevalence of ESBL/AmpC carriage within the pig population (as opposed to the E. coli population) and in 2021, it showed a median prevalence of 54.7%, ranging from 6.5% in Finland to 80.7% in Italy [33]. Comparing these data to the prevalence of ESC resistance in the E. coli population (see Table 1) indicates that, although ESBL/AmpC producers are relatively rare within the general E. coli population, they are widely distributed within the pig population. Some countries voluntarily submit molecular data to the EFSA and ECDC monitoring programme showing that blaCTX-M-1 is the most prevalent ESBL gene in Europe [210], which is in agreement with another recent pan European study [211], monitoring programmes in Denmark [32] and the UK [31], and various other European studies [132,212,213,214,215,216]. The EFSA and ECDC data also showed that in the Netherlands and several of the Nordic countries, mutation of the chromosomal ampC promoter gene was the predominant ESC-resistance mechanism [210], which perhaps reflects a lower prevalence of plasmid-mediated resistance due to the more restrictive AMU regulations in these countries. This was also the case in Denmark [32]. Ewers et al. (2021) examined 99 ESBL/AmpC-producing E. coli isolates recovered from cattle, poultry and pigs at slaughter in eight European countries during the EASSA program [211]. There was marked strain diversity and little evidence of clonality among these isolates, but each ESBL/AmpC gene was associated with particular plasmids, for example, blaCTX-M-1 with IncI1α, many of which were similar to the sequenced plasmids recovered from E. coli in other studies [211]. Although this study included only 15 isolates from pigs (the majority were from poultry), the findings were consistent with several other on-farm studies that showed that ESBL/AmpC genes were distributed across a variety of E. coli strains throughout the farm, as shown in the following examples. Moreover, many of these studies demonstrated that on a given farm, usually one ESBL/AmpC gene and plasmid type predominated. Examples include blaTEM-52 with IncI1, blaCTX-M-1 with IncN, and blaCTX-M-15 with IncFIA/FIB on Portuguese farms in two different studies [217,218]; blaCTX-M-1 with IncN on a Czech farm [219]; blaCTX-M-14 with IncK2 on a Danish farm [220]; and blaCTX-M-1 with IncI on an Australian farm [221]. A longitudinal study that followed pigs from birth until slaughter on 31 Swiss farms had broadly similar findings [222]. These studies demonstrate that plasmids carrying ESBL/Amp genes are widely distributed within the E. coli population but, at least in Europe, are not yet established in the dominant commensal flora. Other studies have demonstrated a high prevalence of ESC resistance in the general E. coli population, for example, one research herd in the USA had a prevalence of resistance to ceftriaxone (associated with blaCMY-2) between 52.1% and 77.1% in weaner pigs [103], and herds using prophylactic antimicrobials in Thailand had a prevalence of resistance of approximately 35% and 45% in finisher pigs in two separate studies [84,128]. Similarly, several studies from China show a high prevalence of resistance to cephalosporins [75,79,80]. Such studies, even if they are not necessarily applicable to the wider population, show that ESBL/AmpC-bearing plasmids have the potential to establish themselves in the dominant E. coli population. As mentioned previously, ESC-resistant E. coli are usually multidrug resistant. Most notably, however, in several of the studies discussed in this section, ESC-resistant E. coli are frequently co-resistant to fluoroquinolones. This phenomenon is particularly well illustrated in the EFSA and ECDC report: while co-resistance is rare in isolates recovered during routine monitoring (i.e., the general E. coli population), 43.3% of the ESC-resistant isolates recovered in specific monitoring of ESBL/AmpC producing E. coli during 2021 were resistant to ciprofloxacin [33]. Hayer et al. (2022) examined over 6000 E. coli genomes and reported a strong association between blaCTX-M and both chromosomal and plasmid-mediated fluoroquinolone-resistant mechanisms [37]. This link between ESC resistance and fluoroquinolone resistance is by no means unique to porcine isolates; it is a notable and concerning problem in human medicine, where, for example, 5.1% of ExPEC isolates submitted to the EARS-Net programme in the EU in 2021 displayed resistance to both classes, along with aminoglycosides [7].
As for studies investigating general antimicrobial resistance in pig production, studies investigating ESC resistance in commensal E. coli mainly involve pigs at or close to slaughter. However, several studies have shown that resistance is highest in younger pigs. There was a higher prevalence of ESC-resistant E. coli carriage in piglets compared with older age groups in four longitudinal studies [119,123,124,223]. Bacterial counts of ESC-resistant E. coli also decreased with age [119,124,223,224,225], although this measurement did not consider the proportion of the population it represents. Other studies in Asia showed a higher prevalence of resistance in weaner pigs [128,226]. Resistance to ESCs has been associated with AMU in some studies. While cephalosporin use at a national level in Europe was not associated with the prevalence of cefotaxime or ceftazidime resistance in the general E. coli population, it was associated with the prevalence of ESBL/AmpC carriage [157]. At farm level, resistance was more likely on farms with cephalosporin use compared with those without use [227,228,229]. Agersø and Aarestrup reported a significant reduction in the prevalence of ESC resistance in Danish slaughter pigs after a voluntary ban on cephalosporin use within the industry [230]. There were similar findings over a four year period on an Australian farm that had ceased cephalosporin use [221].
In comparison with ESC resistance, there are fewer studies investigating fluoroquinolone resistance in the porcine commensal E. coli population. Fluoroquinolones (e.g., ciprofloxacin) have been included in the AST panels in most studies; however, different interpretation criteria and changes in breakpoints over time hamper comparability somewhat. In particular, whether ECOFFs or CBPs are used greatly influences this interpretation as the EUCAST-defined ciprofloxacin cut off MIC for wild type E. coli is currently 0.06 mg/L [42] compared with a CBP (for resistant) of >0.5 mg/L [40,43]. Therefore, studies using ECOFFs typically report a higher prevalence of fluoroquinolone resistance than those using higher breakpoints, as can be seen in the EFSA and ECDC data presented in Table 1. The targets for (fluoro)quinolones are DNA gyrase and topoisomerase IV, which are involved in DNA replication, transcription, and repair. Target modification caused by mutations in the quinolone-resistance determining region (QRDR), usually of gyrA and/or parC, can confer resistance to quinolones, but only low-level resistance to fluoroquinolones [231]. These can be detected if using ECOFFs. Further mutations are required for clinical resistance. Plasmid-mediated quinolone resistance (PMQR) mechanisms such as the qnr family, which protects DNA gyrase and topoisomerase, or genes encoding efflux pumps such as qepA or oxqAB, by themselves confer a low level fluoroquinolone resistance, but facilitate mutations in the QRDR by ‘lengthening’ the mutation prevention window [231,232]. In pigs, chromosomal mutations of gyrA, followed by parC, are the most frequently encountered fluoroquinolone resistance mechanisms while qnrS1 and qnrB19 are the most commonly encountered plasmid-mediated mechanisms [38,68,132,133]. The rates of fluoroquinolone resistance in finisher pigs are typically lower than the rates for the ‘older’ antimicrobials and often higher than ESC resistance (see Table 1 and Table 2). As for ESC resistance, fluoroquinolone resistance is widely distributed in the pig population. In the UK, for example, one cross sectional study found a high level ciprofloxacin resistance on 58% of finisher farms [233], and a more recent WGS-based study found QRDR mutations on 78.5% of farms [132]. Studies investigating fluoroquinolone resistance in younger pigs are scarce. The highest prevalence of fluoroquinolone resistance was found in piglets in an Irish cross-sectional study [65], but was highest in weaner pigs in two smaller-scale longitudinal studies in Vietnam and Brazil [110,226]. Recently, a larger-scale longitudinal study on 24 Swiss farms investigated nalidixic acid resistance in fluoroquinolone-treated and untreated pigs, and found the highest number of positive samples in piglets [120]. Resistance prevalence was also higher in the treated groups and lower on farms not using any fluoroquinolones. Associations between fluoroquinolone use and resistance have also been demonstrated at national- and international level [157,158].
Colistin resistance has become a topic of increasing interest recently. Previously reserved for animal use due to nephrotoxicity in humans, it is now an agent of last resort used to treat some carbapenem-resistant infections in people [234]. Resistance to colistin was previously thought to be chromosomally mediated until the discovery of the plasmid-mediated mcr gene in porcine isolates in China [235]. To date, 10 variants of the mcr gene have been described [236], although the original mcr-1 variant predominates worldwide [38]. While numerous studies have investigated colistin resistance in E. coli of porcine origin [38], only a few have explored the on-farm dynamics. One longitudinal study in Vietnam reported a lower prevalence of colistin resistance in piglets compared with weaners or finishers [226], while a cross-sectional study in Korea showed a higher prevalence of resistance in weaners compared with the older age groups [237]. As seen in Table 1, colistin resistance in E. coli isolated from finishers in the developed world is currently rare; however, as previously discussed, many studies in Asia show a higher prevalence of resistance. In Europe, the prevalence of colistin resistance in pigs at slaughter is associated with the use of colistin at a national level [157].

8. Conclusions and Future Perspectives

In summary, while there is a considerable body of research characterising antimicrobial resistance in porcine E. coli, its relationship with AMU and, to a lesser extent, its evolution during the production cycle, knowledge gaps remain. In particular, longitudinal studies are lacking, especially for fluoroquinolone and colistin resistance. Although the point of slaughter is considered most relevant to human health, AMR in younger pigs, in which resistance is typically higher, is extremely important as this influences AMR at finishing, and is relevant to human occupational exposure and environmental contamination. Thus, more on-farm cross-sectional studies that sample younger pigs or longitudinal studies encompassing the full lifecycle are warranted. The majority of the studies included in this review were conducted in Europe, North America, and, to a lesser extent, China and Southeast Asia. This highlights a lack of data from LMICs and future studies from such countries or regions would be beneficial. In particular, the implementation of integrated monitoring programmes in developing countries is a pressing need as AMU in livestock production is expected to increase in these countries as their food production sectors expand [22]. However, strengthening the surveillance and research of AMR in agriculture is a key component of the Food and Agriculture Organization’s (FAO) action plan on AMR [238], and the successful implementation of such programmes in LMICs will provide essential data to improve the understanding of the epidemiology of AMR in E. coli and other bacteria globally. There is also a need for an increased understanding of the molecular epidemiology of AMR in the general commensal E. coli population as genomic studies, unsurprisingly, tend to focus on the ARGs most important to human health such as ESBL/AmpC, carbapenemases, and mobile resistance genes to fluoroquinolones (e.g., qnr) and polymyxins (i.e., mcr-1). However, the EFSA and ECDC monitoring programme now provides for the use of WGS data [33], and the increasing availability and affordability of WGS is likely to provide intriguing new insights on this topic.
This review was motivated by the potential role played by AMR commensal E. coli of animal origin in the overall epidemiology of AMR. While its overall importance in the overall context of AMR remains to be established, one key question for the future is how will AMR in E. coli evolve in the coming years? On the one hand, several European countries show decreasing trends in resistance to some antimicrobials as their efforts to increase awareness of AMR and improve antimicrobial stewardship take effect [33]. On the other hand, AMU in livestock is projected by some authors to increase in line with the increasing global demand for food [22], and LMICs in Asia have already been identified as ‘hotspots’ for AMR in livestock [37,38,61]. Ultimately, whether this potential threat to human health can be mitigated or reduced depends on the engagement and concerted action of all stakeholders within the health and agriculture sectors. Continued research, monitoring and surveillance will be required into the future to underpin these efforts.

Author Contributions

L.O. performed the literature review and drafted the manuscript. D.E. created the illustration and reviewed and edited the manuscript. L.O., E.G.M. and F.C.L. were responsible for the conceptualisation of the study and reviewed and edited the manuscript. E.G.M. and F.C.L. were responsible for the funding acquisition for the project leading to this publication. All authors have read and agreed to the published version of the manuscript.

Funding

This study was conducted within the AMURAP project (Antimicrobial Use and Resistance in Animal Production) which investigated AMU and AMR in the Irish pig and poultry industries. This project was funded by the Irish Department of Agriculture Food and the Marine (grant reference number 15 S 676).

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABF—antibiotic free; AGISAR—Advisory Group on Integrated Surveillance of Antimicrobial Resistance; AGP—antimicrobial growth promoter; AMR—antimicrobial resistance; AMU—antimicrobial use; APEC—avian pathogenic Escherichia coli; ARG—antimicrobial resistance gene; AST—antimicrobial susceptibility testing; CBP—clinical breakpoint; CIA—critically important antimicrobial; CIPARS—Canadian Integrated Program for Antimicrobial Resistance Surveillance; CLSI—Clinical and Laboratories Standards Institute; CTC—chlortetracycline; DANMAP—Danish Integrated Antimicrobial Resistance Monitoring and Research Programme; DNA—deoxyribonucleic acid; EARS-Net—European Antimicrobial Resistance Surveillance Network; EASSA—European Antimicrobial Susceptibility Surveillance in Animals; ECDC—European Centre for Disease Prevention and Control; ECOFF—epidemiologic cut off value; EEA—European Economic Area; EFFORT—Ecology from Farm to Fork Of microbial drug Resistance and Transmission; EFSA—European Food Safety Agency; EMA—European Medicines Agency; ESBL—extended spectrum beta lactamase; ESC—extended spectrum cephalosporin; ESVAC—European Surveillance of Veterinary Antimicrobial Consumption; ETEC—enterotoxigenic Escherichia coli; EU—European Union; EUCAST—European Committee on Antimicrobial Susceptibility Testing; ExPEC—extra intestinal pathogenic Escherichia coli; FAO—Food and Agriculture Organization; FDA—Food and Drug Administration; HGT—horizontal gene transfer; HP CIA—highest priority critically important antimicrobial; JIACRA—joint inter-agency reports on integrated analysis of antimicrobial agent consumption and occurrence of antimicrobial resistance; JVARM—Japanese Veterinary Antimicrobial Resistance Monitoring System; LMIC—lower and middle income countries; MARAN—Monitoring of Antimicrobial Resistance and Antibiotic Usage in Animals in the Netherlands; MDR—multidrug resistant/multidrug resistance; MGE—mobile genetic element; MIC—minimum inhibitory concentration; NARMS—National Antimicrobial Resistance Monitoring System; PCR—polymerase chain reaction; PMQR—plasmid-mediated fluoroquinolone resistance; QRDR—quinolone resistance determining region; STEC—Shiga toxin producing Escherichia coli; UK—United Kingdom; UK-VARSS—United Kingdom Veterinary Antimicrobial Resistance and Sale Surveillance; USA—United States of America; WGS—whole genome sequencing; WHO—World Health Organization; WT/nWT—wild type/non-wild type.

References

  1. Savageau, M.A. Escherichia coli Habitats, Cell Types, and Molecular Mechanisms of Gene Control. Am. Nat. 1983, 122, 732–744. [Google Scholar] [CrossRef]
  2. Tenaillon, O.; Skurnik, D.; Picard, B.; Denamur, E. The population genetics of commensal Escherichia coli. Nat. Rev. Microbiol. 2010, 8, 207–217. [Google Scholar] [CrossRef]
  3. Kaper, J.B.; Nataro, J.P.; Mobley, H.L. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004, 2, 123–140. [Google Scholar] [CrossRef]
  4. Nataro, J.P.; Kaper, J.B. Diarrheagenic Escherichia coli. Clin. Microbiol. Rev. 1998, 11, 142–201. [Google Scholar] [CrossRef] [PubMed]
  5. European Food Safety Authority (EFSA) and European Centre for Disease Prevention and Control (ECDC). The European Union One Health 2021 Zoonoses Report. EFSA J. 2022, 20, 7666. [Google Scholar] [CrossRef]
  6. Tandogdu, Z.; Wagenlehner, F. Global epidemiology of urinary tract infections. Curr. Opin. Infect. Dis. 2016, 29, 73–79. [Google Scholar] [CrossRef]
  7. European Centre for Disease Prevention and Control (ECDC). Antimicrobial Resistance in the EU/EEA (EARS-Net)—Annual Epidemiological Report. 2021. Available online: https://www.ecdc.europa.eu/en/publications-data/surveillance-antimicrobial-resistance-europe-2021 (accessed on 11 July 2023).
  8. Shane, A.L.; Mody, R.K.; Crump, J.A.; Tarr, P.I.; Steiner, T.S.; Kotloff, K.; Langley, J.M.; Wanke, C.; Warren, C.A.; Cheng, A.C.; et al. 2017 Infectious Diseases Society of America Clinical Practice Guidelines for the Diagnosis and Management of Infectious Diarrhea. Clin. Infect. Dis. 2017, 65, e45–e80. [Google Scholar] [CrossRef] [PubMed]
  9. World Health Organization (WHO). Critically Important Antimicrobials for Human Medicine, 6th Revision. Geneva, Switzerland, 2019; Licence: CC BY-NC-SA 3.0 IGO. Available online: https://www.who.int/publications/i/item/9789241515528 (accessed on 21 July 2021).
  10. World Health Organization (WHO). Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics. 2017. Available online: https://remed.org/wp-content/uploads/2017/03/lobal-priority-list-of-antibiotic-resistant-bacteria-2017.pdf (accessed on 21 September 2023).
  11. Bélanger, L.; Garenaux, A.; Harel, J.; Boulianne, M.; Nadeau, E.; Dozois, C.M. Escherichia coli from Animal Reservoirs as a Potential Source of Human Extraintestinal Pathogenic E. coli. FEMS Immunol. Med. Microbiol. 2011, 62, 1–10. [Google Scholar] [CrossRef]
  12. Manges, A.R.; Johnson, J.R. Food-Borne Origins of Escherichia coli Causing Extraintestinal Infections. Clin. Infect. Dis. 2012, 55, 712–719. [Google Scholar] [CrossRef] [PubMed]
  13. Manges, A.R.; Johnson, J.R. Reservoirs of Extraintestinal Pathogenic Escherichia coli. Microbiol. Spectr. 2015, 3, 159–177. [Google Scholar] [CrossRef]
  14. Jørgensen, S.L.; Stegger, M.; Kudirkiene, E.; Lilje, B.; Poulsen, L.L.; Ronco, T.; Santos, T.; Kiil, K.; Bisgaard, M.; Pedersen, K.; et al. Diversity and Population Overlap between Avian and Human Escherichia coli Belonging to Sequence Type 95. mSphere 2019, 4, e00333-18. [Google Scholar] [CrossRef]
  15. Manges, A.R.; Smith, S.P.; Lau, B.J.; Nuval, C.J.; Eisenberg, J.; Dietrich, P.S.; Riley, L.W. Retail Meat Consumption and the Acquisition of Anti-microbial Resistant Escherichia coli Causing Urinary Tract Infections: A Case Control Study. Foodborne Pathog. Dis. 2007, 4, 419–431. [Google Scholar] [CrossRef]
  16. Manges, A.R. Escherichia coli and urinary tract infections: The role of poultry-meat. Clin. Microbiol. Infect. 2016, 22, 122–129. [Google Scholar] [CrossRef] [PubMed]
  17. Aarestrup, F.M. The livestock reservoir for antimicrobial resistance: A personal view on changing patterns of risks, effects of interventions and the way forward. Phil. Trans. R Soc B. 2015, 370, 20140085. [Google Scholar] [CrossRef]
  18. Winokur, P.; Vonstein, D.; Hoffman, L.; Uhlenhopp, E.; Doe, G. Evidence for Transfer of CMY-2 AmpC β-Lactamase Plasmids between Escherichia coli and Salmonella Isolates from Food Animals and Humans. Antimicrob. Agents Chemother. 2001, 45, 2716–2722. [Google Scholar] [CrossRef]
  19. Mathew, A.G.; Liamthong, S.; Lin, J.; Hong, Y. Evidence of Class 1 Integron Transfer Between Escherichia coli and Salmonella spp. on Livestock Farms. Foodborne Pathog. Dis. 2009, 6, 959–964. [Google Scholar] [CrossRef] [PubMed]
  20. Trobos, M.; Lester, C.H.; Olsen, J.E.; Frimodt-Møller, N.; Hammerum, A.M. Natural transfer of sulphonamide and ampicillin resistance between Escherichia coli residing in the human intestine. J. Antimicrob. Chemoth. 2009, 63, 80–86. [Google Scholar] [CrossRef]
  21. FAOSTAT Crops and Livestocks Products. Available online: https://www.fao.org/faostat/en/#data/TCL (accessed on 2 October 2023).
  22. Mulchandani, R.; Wang, Y.; Gilbert, M.; Boeckel, T.P.V. Global trends in antimicrobial use in food-producing animals: 2020 to 2030. PLoS Glob. Public Health 2023, 3, e0001305. [Google Scholar] [CrossRef] [PubMed]
  23. Ludden, C.; Raven, K.E.; Jamrozy, D.; Gouliouris, T.; Blane, B.; Coll, F.; de Goffau, M.; Naydenova, P.; Horner, C.; Hernandez-Garcia, J.; et al. One Health Genomic Surveillance of Escherichia coli Demonstrates Distinct Lineages and Mobile Genetic Elements in Isolates from Humans versus Livestock. mBio 2019, 10, e02693-18. [Google Scholar] [CrossRef]
  24. Dorado-García, A.; Smid, J.H.; van Pelt, W.; Bonten, M.J.M.J.; Fluit, A.C.; van den Bunt, G.; Wagenaar, J.A.; Hordijk, J.; Dierikx, C.M.; Veldman, K.T.; et al. Molecular relatedness of ESBL/AmpC-producing Escherichia coli from humans, animals, food and the environment: A pooled analysis. J. Antimicrob. Chemother. 2018, 73, 339–347. [Google Scholar] [CrossRef]
  25. Day, M.J.; Hopkins, K.L.; Wareham, D.W.; Toleman, M.A.; Elviss, N.; Randall, L.; Teale, C.; Cleary, P.; Wiuff, C.; Doumith, M.; et al. Extended-spectrum β-lactamase-producing Escherichia coli in human-derived and food chain-derived samples from England, Wales, and Scotland: An epidemiological surveillance and typing study. Lancet Infect. Dis. 2019, 19, 1325–1335. [Google Scholar] [CrossRef]
  26. Mughini-Gras, L.; Dorado-García, A.; van Duijkeren, E.; van den Bunt, G.; Dierikx, C.M.; Bonten, M.J.; Bootsma, M.C.; Schmitt, H.; Hald, T.; Evers, E.G.; et al. Attributable sources of community-acquired carriage of Escherichia coli containing β-lactam antibiotic resistance genes: A population-based modelling study. Lancet Planet Health 2019, 3, e357–e369. [Google Scholar] [CrossRef]
  27. Akwar, T.H.; Poppe, C.; Wilson, J.; Reid-Smith, R.J.; Dyck, M.; Waddington, J.; Shang, D.; Dassie, N.; McEwen, S.A. Risk factors for antimicrobial resistance among fecal Escherichia coli from residents on forty-three swine farms. Microb. Drug Resist. 2007, 13, 69–76. [Google Scholar] [CrossRef] [PubMed]
  28. Dohmen, W.; Bonten, M.J.M.; Bos, M.E.H.; van Marm, S.; Scharringa, J.; Wagenaar, J.A.; Heederik, D.J.J. Carriage of extended-spectrum β-lactamases in pig farmers is associated with occurrence in pigs. Clin. Microbiol. Infect. 2015, 21, 917–923. [Google Scholar] [CrossRef] [PubMed]
  29. Dohmen, W.; Gompel, L.V.; Schmitt, H.; Liakopoulos, A.; Heres, L.; Urlings, B.A.; Mevius, D.; Bonten, M.J.M.; Heederik, D.J.J. ESBL carriage in pig slaughterhouse workers is associated with occupational exposure. Epidemiol. Infect. 2017, 145, 2003–2010. [Google Scholar] [CrossRef]
  30. Dohmen, W.; Liakopoulos, A.; Bonten, M.J.M.; Mevius, D.J.; Heederik, D.J.J. Longitudinal Study of Dynamic Epidemiology of Extended-Spectrum Beta-Lactamase-Producing Escherichia coli in Pigs and Humans Living and/or Working on Pig Farms. Microbiol. Spectr. 2023, 11, e02947-22. [Google Scholar] [CrossRef]
  31. UK-VARSS. UK Veterinary Antibiotic Resistance and Sales Surveillance Report (UK-VARSS 2021). Veterinary Medicines Directorate, Addlestone. 2022. Available online: https://www.gov.uk/government/publications/veterinary-antimicrobial-resistance-and-sales-surveillance-2021 (accessed on 6 January 2023).
  32. DANMAP (Danish Integrated Antimicrobial Resistance Monitoring and Research Programme). DANMAP 2021. Use of Antimicrobial Agents and Occurrence of Antimicrobial Resistance in Bacteria from Food Animals, Food and Humans in Denmark. 2022. Available online: https://www.danmap.org/reports/2021 (accessed on 1 June 2023).
  33. European Food Safety Authority (EFSA) and European Centre for Disease Prevention and Control (ECDC). The European Union Summary Report on Antimicrobial Resistance in Zoonotic and Indicator Bacteria from Humans, Animals and Food in 2020/2021. EFSA J. 2023, 21, 7867. [Google Scholar] [CrossRef]
  34. Luppi, A. Swine enteric colibacillosis: Diagnosis, therapy and antimicrobial resistance. Porc. Health Manag. 2017, 3, 16. [Google Scholar] [CrossRef]
  35. Hayer, S.; Rovira, A.; Olsen, K.; Johnson, T.J.; Vannucci, F.; Rendahl, A.; Perez, A.; Alvarez, J. Prevalence and trend analysis of antimicrobial resistance in clinical Escherichia coli isolates collected from diseased pigs in the USA between 2006 and 2016. Transbound. Emerg. Dis. 2020, 67, 1930–1941. [Google Scholar] [CrossRef]
  36. Burow, E.; Simoneit, C.; Tenhagen, B.A.; Käsbohrer, A. Oral antimicrobials increase antimicrobial resistance in porcine E. coli—A systematic review. Prev. Vet. Med. 2014, 113, 364–375. [Google Scholar] [CrossRef]
  37. Hayer, S.S.; Casanova-Higes, A.; Paladino, E.; Elnekave, E.; Nault, A.; Johnson, T.; Bender, J.; Perez, A.; Alvarez, J. Global Distribution of Extended Spectrum Cephalosporin and Carbapenem Resistance and Associated Resistance Markers in Escherichia coli of Swine Origin—A Systematic Review and Meta-Analysis. Front. Microbiol. 2022, 13, 853810. [Google Scholar] [CrossRef]
  38. Hayer, S.S.; Casanova-Higes, A.; Paladino, E.; Elnekave, E.; Nault, A.; Johnson, T.; Bender, J.; Perez, A.; Alvarez, J. Global Distribution of Fluoroquinolone and Colistin Resistance and Associated Resistance Markers in Escherichia coli of Swine Origin—A Systematic Review and Meta-Analysis. Front. Microbiol. 2022, 13, 834793. [Google Scholar]
  39. Li, M.; Li, Z.; Zhong, Q.; Liu, J.; Han, G.; Li, Y.; Li, C. Antibiotic resistance of fecal carriage of Escherichia coli from pig farms in China: A meta-analysis. Environ. Sci. Pollut. Res. 2022, 29, 22989–23000. [Google Scholar] [CrossRef] [PubMed]
  40. Clinical and Laboratory Standards Institute (CLSI). Performance Standards for Antimicrobial Susceptibility Testing, 33rd ed.; CLSI supplement M100; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2023. [Google Scholar]
  41. Kahlmeter, G.; Turnidge, J. How to: ECOFFs—The why, the how, and the don’ts of EUCAST epidemiological cutoff values. Clin. Microbiol. Infect. 2022, 28, 952–954. [Google Scholar] [CrossRef]
  42. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Data from the EUCAST MIC Distribution Website. 2023. Available online: http://www.eucast.org (accessed on 21 July 2023).
  43. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Breakpoint Tables for Interpretation of MICs and Zone Diameters. Version 13.1. 2023. Available online: https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_13.1_Breakpoint_Tables.pdf (accessed on 4 August 2023).
  44. Kahlmeter, G.; Giske, C.G.; Kirn, T.J.; Sharp, S.E. Point-Counterpoint: Differences between the European Committee on Antimicrobial Susceptibility Testing and Clinical and Laboratory Standards Institute Recommendations for Reporting Antimicrobial Susceptibility Results. J. Clin. Microbiol. 2019, 57, 10–128. [Google Scholar] [CrossRef]
  45. European Committee on Antimicrobial Susceptibility Testing (EUCAST). EUCAST Guidelines for Detection of Resistance Mechanisms and Specific Resistances of Clinical and/or Epidemiological Importance. 2017. Available online: https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Resistance_mechanisms/EUCAST_detection_of_resistance_mechanisms_170711.pdf (accessed on 3 November 2023).
  46. Smith, H.W.; Crabb, W.E. The effect of continuous administration of diets containing low levels of tetracyclines on the incidence of drug-resistant Bacterium coli in the faeces of pigs and chickens: The sensitivity of the Bact. coli to other chemotherapeutic agents. Vet. Rec. 1957, 69, 24–30. [Google Scholar]
  47. Smith, W.H. Antibiotic-resistant Escherichia coli in market pigs in 1956–1979: The emergence of organisms with plasmid-borne trimethoprim resistance. Epidemiol. Infect. 1980, 84, 467–477. [Google Scholar]
  48. Aalbæ, B.; Rasmussen, J.; Nielsen, B.; Olsen, J.E. Prevalence of antibiotic-resistant Escherichia coli in Danish pigs and cattle. APMIS 1991, 99, 1103–1110. [Google Scholar] [CrossRef]
  49. World Health Organization (WHO). Integrated Surveillance of Antimicrobial Resistance in Foodborne Bacteria: Application of a One Health Approach. Geneva, Switzerland, 2017; Licence: CC BY-NC-SA 3.0 IGO. Available online: https://apps.who.int/iris/handle/10665/255747 (accessed on 8 July 2021).
  50. Food and Drug Administration. National Antimicrobial Resistance Monitoring System. Available online: https://www.fda.gov/animal-veterinary/antimicrobial-resistance/national-antimicrobial-resistance-monitoring-system (accessed on 12 August 2023).
  51. Government of Canada. Canadian Integrated Program for Antimicrobial Resistance Surveillance (CIPARS). Available online: https://www.canada.ca/en/public-health/services/surveillance/canadian-integrated-program-antimicrobial-resistance-surveillance-cipars.html (accessed on 12 August 2023).
  52. Ministry of Agriculture, Forestry and Fisheries. Available online: https://www.maff.go.jp/nval/english/AMR/Monitoring/index.html (accessed on 30 September 2023).
  53. de Jong, A.; Thomas, V.; Klein, U.; Marion, H.; Moyaert, H.; Simjee, S.; Vallé, M. Pan-European resistance monitoring programmes encompassing food-borne bacteria and target pathogens of food-producing and companion animals. Int. J. Antimicrob. Agents 2013, 41, 403–409. [Google Scholar] [CrossRef]
  54. DANMAP (Danish Integrated Antimicrobial Resistance Monitoring and Research Programme). Available online: https://www.danmap.org/ (accessed on 30 September 2023).
  55. Wageningen University and Research. Monitoring of Antimicrobial Resistance and Antibiotic Usage in Animals in the Netherlands. Available online: https://www.wur.nl/en/Research-Results/Research-Institutes/Bioveterinary-Research/In-the-spotlight/Antibiotic-resistance/MARAN-reports.htm (accessed on 30 September 2023).
  56. Costa, M.; Cardo, M.; d’Anjo, M.C.; Leite, A. Assessing antimicrobial resistance occurrence in the Portuguese food system: Poultry, pigs and derived food, 2014–2018. Zoonoses Public Health 2022, 69, 312–324. [Google Scholar] [CrossRef]
  57. Duarte, A.S.R.; Marques, A.R.; Andersen, V.D.; Korsgaard, H.B.; Mordhorst, H.; Møller, F.D.; Petersen, T.N.; Vigre, H.; Hald, T.; Aarestrup, F.M. Antimicrobial resistance monitoring in the Danish swine production by phenotypic methods and metagenomics from 1999 to 2018. Eurosurveillance 2023, 28, 2200678. [Google Scholar] [CrossRef]
  58. Sodagari, H.R.; Varga, C. Evaluating Antimicrobial Resistance Trends in Commensal Escherichia coli Isolated from Cecal Samples of Swine at Slaughter in the United States, 2013–2019. Microorganisms 2023, 11, 1033. [Google Scholar] [CrossRef]
  59. Schrijver, R.; Stijntjes, M.; Rodríguez-Baño, J.; Tacconelli, E.; Rajendran, B.N.; Voss, A. Review of antimicrobial resistance surveillance pro-grammes in livestock and meat in EU with focus on humans. Clin. Microbiol. Infect. 2018, 24, 577–590. [Google Scholar] [CrossRef] [PubMed]
  60. Criscuolo, N.G.; Pires, J.; Zhao, C.; Boeckel, T.P.V. resistancebank.org, an open-access repository for surveys of antimicrobial resistance in animals. Sci. Data 2021, 8, 189. [Google Scholar] [CrossRef] [PubMed]
  61. van Boeckel, T.P.; Pires, J.; Silvester, R.; Zhao, C.; Song, J.; Criscuolo, N.G.; Gilbert, M.; Bonhoeffer, S.; Laxminarayan, R. Global trends in antimicrobial resistance in animals in low- and middle-income countries. Science 2019, 365, eaaw1944. [Google Scholar] [CrossRef] [PubMed]
  62. Zhao, C.; Wang, Y.; Tiseo, K.; Pires, J.; Criscuolo, N.G.; van Boeckel, T.P. Geographically targeted surveillance of livestock could help prioritize intervention against antimicrobial resistance in China. Nat. Food 2021, 2, 596–602. [Google Scholar] [CrossRef]
  63. Ramos, S.; Silva, N.; Caniça, M.; Capelo-Martinez, J.; Brito, F.; Igrejas, G.; Poeta, P. High prevalence of antimicrobial-resistant Escherichia coli from animals at slaughter: A food safety risk. J. Sci. Food Agric. 2013, 93, 517–526. [Google Scholar] [CrossRef]
  64. Wasyl, D.; Hoszowski, A.; Zając, M.; Szulowski, K. Antimicrobial resistance in commensal Escherichia coli isolated from animals at slaughter. Front. Microbiol. 2013, 4, 221. [Google Scholar] [CrossRef]
  65. Gibbons, J.; Boland, F.; Egan, J.; Fanning, S.; Markey, B.; Leonard, F. Antimicrobial Resistance of Faecal Escherichia coli Isolates from Pig Farms with Different Durations of In-feed Antimicrobial Use. Zoonoses Public Health 2016, 63, 241–250. [Google Scholar] [CrossRef]
  66. Österberg, J.; Wingstrand, A.; Jensen, A.; Kerouanton, A.; Cibin, V.; Barco, L.; Denis, M.; Aabo, S.; Bengtsson, B. Antibiotic Resistance in Escherichia coli from Pigs in Organic and Conventional Farming in Four European Countries. PLoS ONE 2016, 11, e0157049. [Google Scholar] [CrossRef]
  67. Aasmäe, B.; Häkkinen, L.; Kaart, T.; Kalmus, P. Antimicrobial resistance of Escherichia coli and Enterococcus spp. isolated from Estonian cattle and swine from 2010 to 2015. Acta Vet. Scand. 2019, 61, 5. [Google Scholar] [CrossRef] [PubMed]
  68. Food Drug Administration (FDA) NARMSNow Rockville MD. U.S. Department of Health and Human Services. 2023. Available online: https://www.fda.gov/animal-veterinary/national-antimicrobial-resistance-monitoring-system/narms-now-integrated-data (accessed on 30 September 2023).
  69. Adenipekun, E.O.; Jackson, C.R.; Oluwadun, A.; Iwalokun, B.A.; Frye, J.G.; Barrett, J.B.; Hiott, L.M.; Woodley, T.A. Prevalence and Antimicrobial Resistance in Escherichia coli from Food Animals in Lagos, Nigeria. Microb. Drug Resist. 2015, 21, 358–365. [Google Scholar] [CrossRef]
  70. Ikwap, K.; Gertzell, E.; Hansson, I.; Dahlin, L.; Selling, K.; Magnusson, U.; Dione, M.; Jacobson, M. The presence of antibiotic-resistant Staphylococcus spp. and Escherichia coli in smallholder pig farms in Uganda. BMC Vet. Res. 2021, 17, 31. [Google Scholar] [CrossRef]
  71. Manishimwe, R.; Moncada, P.M.; Musanayire, V.; Shyaka, A.; Scott, M.H.; Loneragan, G.H. Antibiotic-Resistant Escherichia coli and Salmonella from the Feces of Food Animals in the East Province of Rwanda. Animals 2021, 11, 1013. [Google Scholar] [CrossRef] [PubMed]
  72. Katakweba, A.; Muhairwa, A.P.; Lupindu, A.M.; Damborg, P.; Rosenkrantz, J.T.; Minga, U.M.; Mtambo, M.; Olsen, J.E. First Report on a Randomized Investigation of Antimicrobial Resistance in Fecal Indicator Bacteria from Livestock, Poultry, and Humans in Tanzania. Microb. Drug Resist. 2018, 24, 260–268. [Google Scholar] [CrossRef] [PubMed]
  73. Kimera, Z.I.; Mgaya, F.X.; Misinzo, G.; Mshana, S.E.; Moremi, N.; Matee, M.I. Multidrug-Resistant, Including Extended-Spectrum Beta Lactamase-Producing and Quinolone-Resistant, Escherichia coli Isolated from Poultry and Domestic Pigs in Dar es Salaam, Tanzania. Antibiotics 2021, 10, 406. [Google Scholar] [CrossRef]
  74. Fang, J.; Shen, Y.; Qu, D.; Han, J. Antimicrobial resistance profiles and characteristics of integrons in Escherichia coli strains isolated from a large-scale centralized swine slaughterhouse and its downstream markets in Zhejiang, China. Food Control 2019, 95, 215–222. [Google Scholar] [CrossRef]
  75. Zhang, X.; Li, X.; Wang, W.; Qi, J.; Wang, D.; Xu, L.; Liu, Y.; Zhang, Y.; Guo, K. Diverse Gene Cassette Arrays Prevail in Commensal Escherichia coli From Intensive Farming Swine in Four Provinces of China. Front. Microbiol. 2020, 11, 565349. [Google Scholar] [CrossRef]
  76. Lei, T.; Tian, W.; He, L.; Huang, X.H.; Sun, Y.X.; Deng, Y.T.; Sun, Y.; Lv, D.H.; Wu, C.M.; Huang, L.Z.; et al. Antimicrobial resistance in Escherichia coli isolates from food animals, animal food products and companion animals in China. Vet. Microbiol. 2010, 146, 85–89. [Google Scholar] [CrossRef]
  77. Jiang, H.X.; Lü, D.H.; Chen, Z.L.; Wang, X.M.; Chen, J.R.; Liu, Y.H.; Liao, X.P.; Liu, J.H.; Zeng, Z.L. High prevalence and widespread distribution of multi-resistant Escherichia coli isolates in pigs and poultry in China. Vet. J. 2011, 187, 99–103. [Google Scholar] [CrossRef]
  78. Zhang, P.; Shen, Z.; Zhang, C.; Song, L.; Wang, B.; Shang, J.; Yue, X.; Qu, Z.; Li, X.; Wu, L.; et al. Surveillance of antimicrobial resistance among Escherichia coli from chicken and swine, China, 2008–2015. Vet. Microbiol. 2017, 203, 49–55. [Google Scholar] [CrossRef]
  79. Lv, C.; Shang, J.; Zhang, W.; Sun, B.; Li, M.; Guo, C.; Zhou, N.; Guo, X.; Huang, S.; Zhu, Y. Dynamic antimicrobial resistant patterns of Escherichia coli from healthy poultry and swine over 10 years in Chongming Island, Shanghai. Infect. Dis. Poverty 2022, 11, 98. [Google Scholar] [CrossRef] [PubMed]
  80. Ma, J.; Zhou, W.; Wu, J.; Liu, X.; Lin, J.; Ji, X.; Lin, H.; Wang, J.; Jiang, H.; Zhou, Q.; et al. Large-Scale Studies on Antimicrobial Resistance and Molecular Characterization of Escherichia coli from Food Animals in Developed Areas of Eastern China. Microbiol. Spectr. 2022, 10, e02015-22. [Google Scholar] [CrossRef]
  81. Peng, Z.; Hu, Z.; Li, Z.; Zhang, X.; Jia, C.; Li, T.; Dai, M.; Tan, C.; Xu, Z.; Wu, B.; et al. Antimicrobial resistance and population genomics of multidrug-resistant Escherichia coli in pig farms in mainland China. Nat. Commun. 2022, 13, 1116. [Google Scholar] [CrossRef] [PubMed]
  82. Cheng, P.; Yang, Y.; Cao, S.; Liu, H.; Li, X.; Sun, J.; Li, F.; Ishfaq, M.; Zhang, X. Prevalence and Characteristic of Swine-Origin mcr-1-Positive Escherichia coli in Northeastern China. Front. Microbiol. 2021, 12, 712707. [Google Scholar] [CrossRef]
  83. Lugsomya, K.; Chatsuwan, T.; Niyomtham, W.; Tummaruk, P.; Hampson, D.J.; Prapasarakul, N. Routine Prophylactic Antimicrobial Use Is Associated with Increased Phenotypic and Genotypic Resistance in Commensal Escherichia coli Isolates Recovered from Healthy Fattening Pigs on Farms in Thailand. Microb. Drug Resist. 2018, 24, 213–223. [Google Scholar] [CrossRef] [PubMed]
  84. Pholwat, S.; Pongpan, T.; Chinli, R.; McQuade, E.T.; Thaipisuttikul, I.; Ratanakorn, P.; Liu, J.; Taniuchi, M.; Houpt, E.R.; Foongladda, S. Antimicrobial Resistance in Swine Fecal Specimens Across Different Farm Management Systems. Front. Microbiol. 2020, 11, 1238. [Google Scholar] [CrossRef]
  85. Trongjit, S.; Angkittitrakul, S.; Chuanchuen, R. Occurrence and molecular characteristics of antimicrobial resistance of Escherichia coli from broilers, pigs and meat products in Thailand and Cambodia provinces. Microbiol. Immunol. 2016, 60, 575–585. [Google Scholar] [CrossRef]
  86. Tuat, C.V.; Hue, P.T.; Loan, N.T.P.; Thuy, N.T.; Hue, L.T.; Giang, V.N.; Erickson, V.I.; Padungtod, P. Antimicrobial Resistance Pilot Surveillance of Pigs and Chickens in Vietnam, 2017–2019. Front. Vet. Sci. 2021, 8, 618497. [Google Scholar] [CrossRef]
  87. Government of Canada. Canadian Integrated Program for Antimicrobial Resistance Surveillance (CIPARS) 2019: Figures and Tables. Public Health Agency of Canada, Guelph. 2020. Available online: https://publications.gc.ca/collections/collection_2022/aspc-phac/HP2-4-2019-eng-5.pdf (accessed on 4 November 2023).
  88. Ministry of Agriculture, Forestry and Fisheries. Report on the Japanese Veterinary Antimicrobial Resistance Monitoring System 2016–2017. 2020. Available online: https://www.maff.go.jp/nval/yakuzai/pdf/200731_JVARMReport_2016–2017.pdf (accessed on 20 July 2021).
  89. Teshager, T.; Herrero, I.A.; Porrero, M.; Garde, J.; Moreno, M.A.; Domínguez, L. Surveillance of antimicrobial resistance in Escherichia coli strains isolated from pigs at Spanish slaughterhouses. Int. J. Antimicrob. Agents 2000, 15, 137–142. [Google Scholar] [CrossRef]
  90. Sáenz, Y.; Zarazaga, M.; Briñas, L.; Lantero, M.; Ruiz-Larrea, F.; Torres, C. Antibiotic resistance in Escherichia coli isolates obtained from animals, foods and humans in Spain. Int. J. Antimicrob. Agents 2001, 18, 353–358. [Google Scholar] [CrossRef] [PubMed]
  91. de Jong, A.; Thomas, V.; Simjee, S.; Godinho, K.; Schiessl, B.; Klein, U.; Butty, P.; Vallé, M.; Marion, H.; Shryock, T.R. Pan-European monitoring of susceptibility to human-use antimicrobial agents in enteric bacteria isolated from healthy food-producing animals. J. Antimicrob. Chemoth. 2012, 67, 638–651. [Google Scholar] [CrossRef] [PubMed]
  92. de Jong, A.; Garch, F.E.; Hocquet, D.; Prenger-Berninghoff, E.; Dewulf, J.; Migura-Garcia, L.; Perrin-Guyomard, A.; Veldman, K.T.; Janosi, S.; Skarzynska, M.; et al. European-wide antimicrobial resistance monitoring in commensal Escherichia coli isolated from healthy food animals between 2004 and 2018. J. Antimicrob. Chemother. 2022, 77, 3301–3311. [Google Scholar] [CrossRef] [PubMed]
  93. Dunlop, R.H.; McEwen, S.A.; Meek, A.H.; Black, W.D.; Friendship, R.M.; Clarke, R.C. Prevalences of resistance to seven antimicrobials among fecal Escherichia coli of swine on thirty-four farrow-to-finish farms in Ontario, Canada. Prev. Vet. Med. 1998, 34, 265–282. [Google Scholar] [CrossRef] [PubMed]
  94. Varga, C.; Rajić, A.; McFall, M.E.; Avery, B.P.; Reid-Smith, R.J.; Deckert, A.; Checkley, S.L.; McEwen, S.A. Antimicrobial resistance in generic Escherichia coli isolated from swine fecal samples in 90 Alberta finishing farms. Can. J. Vet. Res. 2008, 72, 175–180. [Google Scholar] [PubMed]
  95. Rosengren, L.B.; Waldner, C.L.; Reid-Smith, R.J.; Checkley, S.L.; McFall, M.E.; Rajić, A. Antimicrobial resistance of fecal Escherichia coli isolated from grow-finish pigs in 20 herds in Alberta and Saskatchewan. Can. J. Vet. Res. 2008, 72, 160–167. [Google Scholar]
  96. Akwar, H.; Poppe, C.; Wilson, J.; Reid-Smith, R.J.; Dyck, M.; Waddington, J.; Shang, D.; McEwen, S.A. Prevalence and patterns of antimicrobial resistance of fecal Escherichia coli among pigs on 47 farrow-to-finish farms with different in-feed medication policies. Can. J. Vet. Res. 2008, 72, 195–201. [Google Scholar]
  97. Bunner, C.A.; Norby, B.; Bartlett, P.C.; Erskine, R.J.; Downes, F.P.; Kaneene, J.B. Prevalence and pattern of antimicrobial susceptibility in Escherichia coli isolated from pigs reared under antimicrobial-free and conventional production methods. J. Am. Vet. Med. Assoc. 2007, 231, 275–283. [Google Scholar] [CrossRef]
  98. Smith, M.; Jordan, D.; Gibson, J.; Cobbold Chapman, T.; Abraham, S.; Trott, D. Phenotypic and genotypic profiling of antimicrobial resistance in enteric Escherichia coli communities isolated from finisher pigs in Australia. Aust. Vet. J. 2016, 94, 371–376. [Google Scholar] [CrossRef]
  99. Kidsley, A.K.; Abraham, S.; Bell, J.M.; O’Dea, M.; Laird, T.J.; Jordan, D.; Mitchell, P.; McDevitt, C.A.; Trott, D.J. Antimicrobial Susceptibility of Escherichia coli and Salmonella spp. Isolates From Healthy Pigs in Australia: Results of a Pilot National Survey. Front. Microbiol. 2018, 9, 1207. [Google Scholar] [CrossRef]
  100. Lim, S.K.; Lee, H.S.; Nam, H.M.; Cho, Y.S.; Kim, J.M.; Song, S.W.; Park, Y.H.; Jung, S.C. Antimicrobial resistance observed in Escherichia coli strains isolated from fecal samples of cattle and pigs in Korea during 2003–2004. Int. J. Food Microbiol. 2007, 116, 283–286. [Google Scholar] [CrossRef]
  101. Lee, M.; Shin, E.; Lee, Y. Antimicrobial Resistance and Integron Profiles in Multidrug-Resistant Escherichia coli Isolates from Pigs. Foodborne Pathog. Dis. 2014, 11, 988–997. [Google Scholar] [CrossRef]
  102. Song, H.J.; Kim, S.J.; Moon, D.C.; Mechesso, A.F.; Choi, J.H.; Kang, H.Y.; Boby, N.; Yoon, S.S.; Lim, S.K. Antimicrobial Resistance in Escherichia coli Isolates from Healthy Food Animals in South Korea, 2010–2020. Microorganisms 2022, 10, 524. [Google Scholar] [CrossRef]
  103. Agga, G.; Scott, H.; Amachawadi, R.; Nagaraja, T.; Vinasco, J.; Bai, J.; Norby, B.; Renter, D.; Dritz, S.; Nelssen, J.; et al. Effects of chlortetracycline and copper supplementation on antimicrobial resistance of fecal Escherichia coli from weaned pigs. Prev. Vet. Med. 2014, 114, 231–246. [Google Scholar] [CrossRef]
  104. Bibbal, D.; Dupouy, V.; Ferré, J.; Toutain, P.; Fayet, O.; Prère, M.; Bousquet-Mélou, A. Impact of Three Ampicillin Dosage Regimens on Selection of Ampicillin Resistance in Enterobacteriaceae and Excretion of blaTEM Genes in Swine Feces. Appl. Environ. Microbiol. 2007, 73, 4785–4790. [Google Scholar] [CrossRef]
  105. Gaire, T.N.; Scott, H.M.; Sellers, L.; Nagaraja, T.G.; Volkova, V.V. Age Dependence of Antimicrobial Resistance Among Fecal Bacteria in Animals: A Scoping Review. Front. Vet. Sci. 2021, 7, 622495. [Google Scholar] [CrossRef]
  106. Lekagul, A.; Tangcharoensathien, V.; Yeung, S. Patterns of antibiotic use in global pig production: A systematic review. Vet. Anim. Sci. 2019, 7, 00058. [Google Scholar] [CrossRef] [PubMed]
  107. Sarrazin, S.; Joosten, P.; Van Gompel, L.; Luiken, R.E.; Mevius, D.J.; Wagenaar, J.A.; Heederik, D.J.; Dewulf, J. Quantitative and qualitative analysis of antimicrobial usage patterns in 180 selected farrow-to-finish pig farms from nine European countries based on single batch and purchase data. J. Antimicrob. Chemother. 2019, 74, 807–816. [Google Scholar] [CrossRef] [PubMed]
  108. Alali, W.Q.; Scott, H.M.; Harvey, R.B.; Norby, B.; Lawhorn, D.B.; Pillai, S.D. Longitudinal Study of Antimicrobial Resistance among Escherichia coli Isolates from Integrated Multisite Cohorts of Humans and Swine. Appl. Environ. Microbiol. 2008, 74, 3672–3681. [Google Scholar] [CrossRef] [PubMed]
  109. Marchant, M.; Moreno, M.A. Dynamics and Diversity of Escherichia coli in Animals and System Management of the Manure on a Commercial Farrow-to-Finish Pig Farm. Appl. Environ. Microbiol. 2013, 79, 853–859. [Google Scholar] [CrossRef] [PubMed]
  110. Pissetti, C.; Kich, J.; Allen, H.K.; Navarrete, C.; de Costa, E.; Morés, N.; Cardoso, M. Antimicrobial resistance in commensal Escherichia coli and Enterococcus spp. isolated from pigs subjected to different antimicrobial administration protocols. Res. Vet. Sci. 2021, 137, 174–185. [Google Scholar] [CrossRef] [PubMed]
  111. Langlois, B.; Dawson, K.; Leak, I.; Aaron, D. Effect of age and housing location on antibiotic resistance of fecal coliforms from pigs in a non-antibiotic-exposed herd. Appl. Environ. Microbiol. 1988, 54, 1341–1344. [Google Scholar] [CrossRef]
  112. Yun, J.; Muurinen, J.; Nykäsenoja, S.; Seppä-Lassila, L.; Sali, V.; Suomi, J.; Tuominen, P.; Joutsen, S.; Hämäläinen, M.; Olkkola, S.; et al. Antimicrobial use, biosecurity, herd characteristics, and antimicrobial resistance in indicator Escherichia coli in ten Finnish pig farms. Prev. Vet. Med. 2021, 193, 105408. [Google Scholar] [CrossRef] [PubMed]
  113. Burow, E.; Rostalski, A.; Harlizius, J.; Gangl, A.; Simoneit, C.; Grobbel, M.; Kollas, C.; Tenhagen, B.A.A.; Käsbohrer, A. Antibiotic resistance in Escherichia coli from pigs from birth to slaughter and its association with antibiotic treatment. Prev. Vet. Med. 2019, 165, 52–62. [Google Scholar] [CrossRef] [PubMed]
  114. Mathew, A.; Beckmann, M.; Saxton, A.M. A comparison of antibiotic resistance in bacteria isolated from swine herds in which antibiotics were used or excluded. J. Swine Health Prod. 2001, 9, 125–129. [Google Scholar]
  115. Græsbøll, K.; Damborg, P.; Mellerup, A.; Herrero-Fresno, A.; Larsen, I.; Holm, A.; Nielsen, J.P.; Christiansen, L.E.; Angen, Ø.; Ahmed, S.; et al. Effect of Tetracycline Dose and Treatment Mode on Selection of Resistant Coliform Bacteria in Nursery Pigs. Appl. Environ. Microbiol. 2017, 83, e00538-17. [Google Scholar] [CrossRef] [PubMed]
  116. Katouli, M.; Lund, A.; Wallgren, P.; Kühn, I.; Söderlind, O.; Möllby, R. Phenotypic characterization of intestinal Escherichia coli of pigs during suckling, postweaning, and fattening periods. Appl. Environ. Microbiol. 1995, 61, 778–783. [Google Scholar] [CrossRef]
  117. Schierack, P.; Kadlec, K.; Guenther, S.; Filter, M.; Schwarz, S.; Ewers, C.; Wieler, L.H. Antimicrobial resistances do not affect colonization parameters of intestinal E. coli in a small piglet group. Gut Pathog. 2009, 1, 18. [Google Scholar] [CrossRef]
  118. Ahmed, S.; Olsen, J.E.; Herrero-Fresno, A. The genetic diversity of commensal Escherichia coli strains isolated from non-antimicrobial treated pigs varies according to age group. PLoS ONE 2017, 12, e0178623. [Google Scholar] [CrossRef]
  119. Hansen, K.; Damborg, P.; Andreasen, M.; Nielsen, S.; Guardabassi, L. Carriage and Fecal Counts of Cefotaxime M-Producing Escherichia coli in Pigs: A Longitudinal Study. Appl. Environ. Microbiol. 2013, 79, 794–798. [Google Scholar] [CrossRef]
  120. Amsler, M.; Zurfluh, K.; Hartnack, S.; Sidler, X.; Stephan, R.; Kümmerlen, D. Occurrence of Escherichia coli non-susceptible to quinolones in faecal samples from fluoroquinolone-treated, contact and control pigs of different ages from 24 Swiss pig farms. Porc. Health Manag. 2021, 7, 29. [Google Scholar] [CrossRef] [PubMed]
  121. Mathew, A.G.; Garner, K.N.; Ebner, P.D.; Saxton, A.M.; Clift, R.E.; Liamthong, S. Effects of Antibiotic Use in Sows on Resistance of E. coli and Salmonella enterica Typhimurium in Their Offspring. Foodborne Pathog. Dis. 2005, 2, 212–220. [Google Scholar] [CrossRef] [PubMed]
  122. Callens, B.; Faes, C.; Maes, D.; Catry, B.; Boyen, F.; Francoys, D.; de Jong, E.; Haesebrouck, F.; Dewulf, J. Presence of Antimicrobial Resistance and Antimicrobial Use in Sows Are Risk Factors for Antimicrobial Resistance in Their Offspring. Microb. Drug Resist. 2015, 21, 50–58. [Google Scholar] [CrossRef]
  123. Cameron-Veas, K.; Solà-Ginés, M.; Moreno, M.A.; Fraile, L.; Migura-Garcia, L. Impact of the Use of β-Lactam Antimicrobials on the Emergence of Escherichia coli Isolates Resistant to Cephalosporins under Standard Pig-Rearing Conditions. Appl. Environ. Microbiol. 2015, 81, 1782–1787. [Google Scholar] [CrossRef]
  124. Cameron-Veas, K.; Moreno, M.A.; Fraile, L.; Migura-Garcia, L. Shedding of cephalosporin resistant Escherichia coli in pigs from conventional farms after early treatment with antimicrobials. Vet. J. 2016, 211, 21–25. [Google Scholar] [CrossRef]
  125. Stannarius, C.; Bürgi, E.; Regula, G.; Zychowska, M.; Zweifel, C.; Stephan, R. Antimicrobial resistance in Escherichia coli strains isolated from Swiss weaned pigs and sows. Schweiz. Arch. Für Tierheilkd. 2009, 151, 119–125. [Google Scholar] [CrossRef]
  126. Hallenberg, G.; Jiwakanon, J.; Angkititrakul, S.; Kangair, S.; Osbjer, K.; Lunha, K.; Sunde, M.; Järhult, J.D.; Boeckel, T.P.; Rich, K.M.; et al. Antibiotic use in pig farms at different levels of intensification—Farmers’ practices in northeastern Thailand. PLoS ONE 2020, 15, e0243099. [Google Scholar] [CrossRef] [PubMed]
  127. Lunha, K.; Leangapichart, T.; Jiwakanon, J.; Angkititrakul, S.; Sunde, M.; Järhult, J.D.; Hallenberg, G.; Hickman, R.A.; Boeckel, T.; Magnusson, U. Antimicrobial Resistance in Fecal Escherichia coli from Humans and Pigs at Farms at Different Levels of Intensification. Antibiotics 2020, 9, 662. [Google Scholar] [CrossRef]
  128. Kittitat, L.; Jitrapa, Y.; Waree, N.; Chanwit, T.; Padet, T.; Hampson, D.J.; Nuvee, P. Antimicrobial Resistance in Commensal Escherichia coli Isolated from Pigs and Pork Derived from Farms Either Routinely Using or Not Using in-Feed Antimicrobials. Microb. Drug Resist. 2018, 24, 1054–1066. [Google Scholar]
  129. Sodagari, H.R.; Agrawal, I.; Yudhanto, S.; Varga, C. Longitudinal analysis of differences and similarities in antimicrobial resistance among commensal Escherichia coli isolated from market swine and sows at slaughter in the United States of America, 2013–2019. Int. J. Food Microbiol. 2023, 407, 110388. [Google Scholar] [CrossRef]
  130. Leekitcharoenphon, P.; Johansson, M.; Munk, P.; Malorny, B.; Skarżyńska, M.; Wadepohl, K.; Moyano, G.; Hesp, A.; Veldman, K.T.; Bossers, A.; et al. Genomic evolution of antimicrobial resistance in Escherichia coli. Sci. Rep. 2021, 11, 15108. [Google Scholar] [CrossRef] [PubMed]
  131. Stubberfield, E.; AbuOun, M.; Sayers, E.; O’Connor, H.M.; Card, R.M.; Anjum, M.F. Use of whole genome sequencing of commensal Escherichia coli in pigs for antimicrobial resistance surveillance, United Kingdom, 2018. Eurosurveillance 2019, 24, 1900136. [Google Scholar] [CrossRef]
  132. AbuOun, M.; O’Connor, H.M.; Stubberfield, E.J.; Nunez-Garcia, J.; Sayers, E.; Crook, D.W.; Smith, R.P.; Anjum, M.F. Characterizing Antimicrobial Resistant Escherichia coli and Associated Risk Factors in a Cross-Sectional Study of Pig Farms in Great Britain. Front. Microbiol. 2020, 11, 861. [Google Scholar] [CrossRef]
  133. Mencía-Ares, O.; Borowiak, M.; Argüello, H.; Cobo-Díaz, J.F.; Malorny, B.; Álvarez-Ordóñez, A.; Carvajal, A.; Deneke, C. Genomic Insights into the Mobilome and Resistome of Sentinel Microorganisms Originating from Farms of Two Different Swine Production Systems. Microbiol. Spectr. 2022, 10, e02896-22. [Google Scholar] [CrossRef] [PubMed]
  134. Reid, C.J.; Wyrsch, E.R.; Chowdhury, P.R.; Zingali, T.; Liu, M.; Darling, A.E.; Chapman, T.A.; Djordjevic, S.P. Porcine commensal Escherichia coli: A reservoir for class 1 integrons associated with IS26. Microb. Genom. 2017, 3, e000143. [Google Scholar] [CrossRef]
  135. Zingali, T.; Reid, C.J.; Chapman, T.A.; Gaio, D.; Liu, M.; Darling, A.E.; Djordjevic, S.P. Whole Genome Sequencing Analysis of Porcine Faecal Commensal Escherichia coli Carrying Class 1 Integrons from Sows and Their Offspring. Microorganisms 2020, 8, 843. [Google Scholar] [CrossRef] [PubMed]
  136. Pires, J.; Huisman, J.S.; Bonhoeffer, S.; Boeckel, T.P.V. Increase in antimicrobial resistance in Escherichia coli in food animals between 1980 and 2018 assessed using genomes from public databases. J. Antimicrob. Chemother. 2021, 77, 646–655. [Google Scholar] [CrossRef]
  137. Poirel, L.; Madec, J.Y.Y.; Lupo, A.; Schink, A.K.K.; Kieffer, N.; Nordmann, P.; Schwarz, S. Antimicrobial Resistance in Escherichia coli. Microbiol. Spectr. 2018, 6, 6-4. [Google Scholar] [CrossRef]
  138. Jurado-Rabadán, S.; de la Fuente, R.; Ruiz-Santa-Quiteria, J.A.; Orden, J.A.; de Vries, L.E.; Agersø, Y. Detection and linkage to mobile genetic elements of tetracycline resistance gene tet(M) in Escherichia coli isolates from pigs. BMC Vet. Res. 2014, 10, 155. [Google Scholar] [CrossRef]
  139. Enne, V.I.; Cassar, C.; Sprigings, K.; Woodward, M.J.; Bennett, P.M. A high prevalence of antimicrobial resistant Escherichia coli isolated from pigs and a low prevalence of antimicrobial resistant E. coli from cattle and sheep in Great Britain at slaughter. FEMS Microbiol. Lett. 2008, 278, 193–199. [Google Scholar] [CrossRef]
  140. Schwarz, S.; Kehrenberg, C.; Doublet, B.; Cloeckaert, A. Molecular basis of bacterial resistance to chloramphenicol and florfenicol. FEMS Microbiol. Lett. 2004, 28, 519–542. [Google Scholar] [CrossRef]
  141. Du, Z.; Wang, M.; Cui, G.; Zu, X.; Zhao, Z.; Xue, Y. The prevalence of amphenicol resistance in Escherichia coli isolated from pigs in main-land China from 2000 to 2018: A systematic review and meta-analysis. PLoS ONE 2020, 15, e0228388. [Google Scholar] [CrossRef] [PubMed]
  142. Hall, M.A.; Blok, H.E.; Donders, R.A.; Paauw, A.; Fluit, A.C.; Verhoef, J. Multidrug Resistance among Enterobacteriaceae Is Strongly Associated with the Presence of Integrons and Is Independent of Species or Isolate Origin. J. Infect. Dis. 2003, 187, 251–259. [Google Scholar]
  143. Hall, R.M.; Collis, C.M. Mobile gene cassettes and integrons: Capture and spread of genes by site-specific recombination. Mol. Microbiol. 1995, 15, 593–600. [Google Scholar] [CrossRef] [PubMed]
  144. Domingues, S.; da Silva, G.J.; Nielsen, K.M. Integrons: Vehicles and pathways for horizontal dissemination in bacteria. Mobile genetic elements. Mob. Genet. Elem. 2012, 2, 211–223. [Google Scholar] [CrossRef] [PubMed]
  145. Bischoff, K.M.; White, D.G.; Hume, M.E.; Poole, T.L.; Nisbet, D.J. The chloramphenicol resistance gene cmlA is disseminated on transferable plasmids that confer multiple-drug resistance in swine Escherichia coli. FEMS Microbiol. Lett. 2005, 243, 285–291. [Google Scholar] [CrossRef]
  146. Dawes, F.E.; Kuzevski, A.; Bettelheim, K.A.; Hornitzky, M.A.; Djordjevic, S.P.; Walker, M.J. Distribution of Class 1 Integrons with IS26-Mediated Deletions in Their 3′-Conserved Segments in Escherichia coli of Human and Animal Origin. PLoS ONE 2010, 5, e12754. [Google Scholar] [CrossRef]
  147. Marchant, M.; Vinué, L.; Torres, C.; Moreno, M.A. Change of integrons over time in Escherichia coli isolates recovered from healthy pigs and chickens. Vet. Microbiol. 2013, 163, 124–132. [Google Scholar] [CrossRef]
  148. Changkaew, K.; Intarapuk, A.; Utrarachkij, F.; Nakajima, C.; Suthienkul, O.; Suzuki, Y. Antimicrobial Resistance, Extended-Spectrum β-Lactamase Productivity, and Class 1 Integrons in Escherichia coli from Healthy Swine. J. Food Protect. 2016, 78, 1442–1450. [Google Scholar] [CrossRef]
  149. Chaslus-Dancla, E.; Pohl, P.; Meurisse, M.; Marin, M.; Lafont, J. High genetic homology between plasmids of human and animal origins conferring resistance to the aminoglycosides gentamicin and apramycin. Antimicrob. Agents Chemother. 1991, 35, 590–593. [Google Scholar] [CrossRef]
  150. Jensen, V.F.; Jakobsen, L.; Emborg, H.D.; Seyfarth, A.; Hammerum, A.M. Correlation between apramycin and gentamicin use in pigs and an increasing reservoir of gentamicin-resistant Escherichia coli. J. Antimicrob. Chemother. 2006, 58, 101–107. [Google Scholar] [CrossRef] [PubMed]
  151. Choi, M.J.; Lim, S.K.; Nam, H.M.; Kim, A.R.; Jung, S.C.; Kim, M.N. Apramycin and Gentamicin Resistances in Indicator and Clinical Escherichia coli Isolates from Farm Animals in Korea. Foodborne Pathog Dis. 2011, 8, 119–123. [Google Scholar] [CrossRef] [PubMed]
  152. Pankey, G.A. Tigecycline. J. Antimicrob. Chemother. 2005, 56, 470–480. [Google Scholar] [CrossRef] [PubMed]
  153. He, T.; Wang, R.; Liu, D.; Walsh, T.R.; Zhang, R.; Lv, Y.; Ke, Y.; Ji, Q.; Wei, R.; Liu, Z.; et al. Emergence of plasmid-mediated high-level tigecycline resistance genes in animals and humans. Nat. Microbiol. 2019, 4, 1450–1456. [Google Scholar] [CrossRef] [PubMed]
  154. Chantziaras, I.; Boyen, F.; Callens, B.; Dewulf, J. Correlation between veterinary antimicrobial use and antimicrobial resistance in food-producing animals: A report on seven countries. J. Antimicrob. Chemother. 2014, 69, 827–834. [Google Scholar] [CrossRef]
  155. Callens, B.; Cargnel, M.; Sarrazin, S.; Dewulf, J.; Hoet, B.; Vermeersch, K.; Wattiau, P.; Welby, S. Associations between a decreased veterinary antimicrobial use and resistance in commensal Escherichia coli from Belgian livestock species (2011–2015). Prev. Vet. Med. 2018, 157, 50–58. [Google Scholar] [CrossRef]
  156. Asai, T.; Kojima, A.; Harada, K.; Ishihara, K.; Takahashi, T.; Tamura, Y. Correlation between the usage volume of veterinary therapeutic antimicrobials and resistance in Escherichia coli isolated from the feces of food-producing animals in Japan. Jpn J. Infect. Dis. 2005, 58, 369–372. [Google Scholar]
  157. European Centre for Disease Prevention and Control (ECDC); European Food Safety Authority (EFSA); European Medicines Agency (EMA). Third joint inter-agency report on integrated analysis of consumption of antimicrobial agents and occurrence of antimicrobial resistance in bacteria from humans and food-producing animals in the EU/EEA. EFSA J. 2021, 19, e06712. [Google Scholar] [CrossRef]
  158. Ceccarelli, D.; Hesp, A.; van der Goot, J.; Joosten, P.; Sarrazin, S.; Wagenaar, J.A.; Dewulf, J.; Mevius, D.J.; Consortium, O. Antimicrobial resistance prevalence in commensal Escherichia coli from broilers, fattening turkeys, fattening pigs and veal calves in European countries and association with antimicrobial usage at country level. J. Med. Microbiol. 2020, 69, 537–547. [Google Scholar] [CrossRef]
  159. Dorado-García, A.; Mevius, D.J.; Jacobs, J.J.; Geijlswijk, I.M.; Mouton, J.W.; Wagenaar, J.A.; Heederik, D.J. Quantitative assessment of antimicrobial resistance in livestock during the course of a nationwide antimicrobial use reduction in the Netherlands. J. Antimicrob. Chemother. 2016, 71, 3607–3619. [Google Scholar] [CrossRef]
  160. Wang, Y.; Xu, C.; Zhang, R.; Chen, Y.; Shen, Y.; Hu, F.; Liu, D.; Lu, J.; Guo, Y.; Xia, X.; et al. Changes in colistin resistance and mcr-1 abundance in Escherichia coli of animal and human origins following the ban of colistin-positive additives in China: An epidemiological comparative study. Lancet Infect. Dis. 2020, 20, 1161–1171. [Google Scholar] [CrossRef] [PubMed]
  161. Shen, C.; Zhong, L.L.; Yang, Y.; Doi, Y.; Paterson, D.L.; Stoesser, N.; Ma, F.; Ahmed, M.A.E.G.E.S.; Feng, S.; Huang, S.; et al. Dynamics of mcr-1 prevalence and mcr-1-positive Escherichia coli after the cessation of colistin use as a feed additive for animals in China: A prospective cross-sectional and whole genome sequencing-based molecular epidemiological study. Lancet Microbe 2020, 1, e34–e43. [Google Scholar] [CrossRef] [PubMed]
  162. Langlois, B.; Cromwell, G.; Hays, V. Influence of Type of Antibiotic and Length of Antibiotic Feeding Period on Performance and Persistence of Antibiotic Resistant Enteric Bacteria in Growing-Finishing Swine. J. Anim. Sci. 1978, 46, 1383–1396. [Google Scholar] [CrossRef]
  163. Langlois, B.; Dawson, K.; Stahly, T.; Cromwell, G. Antibiotic Resistance of Fecal Coliforms from Swine Fed Subtherapeutic and Therapeutic Levels of Chlortetracycline. J. Anim. Sci. 1984, 58, 666–674. [Google Scholar] [CrossRef] [PubMed]
  164. Delsol, A.A.; Anjum, M.; Woodward, M.J.; Sunderland, J.; Roe, J.M. The effect of chlortetracycline treatment and its subsequent withdrawal on multi-resistant Salmonella enterica serovar Typhimurium DT104 and commensal Escherichia coli in the pig. J. Appl. Microbiol. 2003, 95, 1226–1234. [Google Scholar] [CrossRef]
  165. Funk, J.A.; Lejeune, J.T.; Wittum, T.E.; Rajala-Schultz, P.J. The Effect of Subtherapeutic Chlortetracycline on Antimicrobial Resistance in the Fecal Flora of Swine. Microb. Drug Resist. 2006, 12, 210–218. [Google Scholar] [CrossRef]
  166. Mathew, A.G.; Arnett, D.B.; Cullen, P.; Ebner, P.D. Characterization of resistance patterns and detection of apramycin resistance genes in Escherichia coli isolated from swine exposed to various environmental conditions. Int. J. Food Microbiol. 2003, 89, 11–20. [Google Scholar] [CrossRef]
  167. Moro, M.; Beran, G.; Hoffman, L.; Griffith, R. Effects of cold stress on the antimicrobial drug resistance of Escherichia coli of the intestinal flora of swine. Lett. Appl. Microbiol. 1998, 27, 251–254. [Google Scholar] [CrossRef]
  168. Moro, M.H.; Beran, G.W.; Griffith, R.W.; Hoffman, L.J. Effects of heat stress on the antimicrobial drug resistance of Escherichia coli of the intestinal flora of swine. J. Appl. Microbiol. 2000, 88, 836–844. [Google Scholar] [CrossRef]
  169. Mathew, A.; Jackson, F.; Saxton, A.M. Effects of antibiotic regimens on resistance of Escherichia coli and Salmonella serovar Typhimurium in swine. J. Swine Health Prod. 2002, 10, 7–13. [Google Scholar]
  170. Herrero-Fresno, A.; Zachariasen, C.; Nørholm, N.; Holm, A.; Christiansen, L.; Olsen, J. Effect of different oral oxytetracycline treatment regimes on selection of antimicrobial resistant coliforms in nursery pigs. Vet. Microbiol. 2017, 208, 1–7. [Google Scholar] [CrossRef] [PubMed]
  171. Bibbal, D.; Dupouy, V.; Prère, M.; Toutain, P.; Bousquet-Mélou, A. Relatedness of Escherichia coli Strains with Different Susceptibility Phenotypes Isolated from Swine Feces during Ampicillin Treatment. Appl. Environ. Microbiol. 2009, 75, 2999–3006. [Google Scholar] [CrossRef]
  172. Herrero-Fresno, A.; Zachariasen, C.; Hansen, M.; Nielsen, A.; Hendriksen, R.S.; Nielsen, S.; Olsen, J. Apramycin treatment affects selection and spread of a multidrug-resistant Escherichia coli strain able to colonize the human gut in the intestinal microbiota of pigs. Vet. Res. 2015, 47, 12. [Google Scholar] [CrossRef]
  173. Römer, A.; Scherz, G.; Reupke, S.; Meißner, J.; Wallmann, J.; Kietzmann, M.; Kaspar, H. Effects of intramuscularly administered enrofloxacin on the susceptibility of commensal intestinal Escherichia coli in pigs (Sus scrofa domestica). BMC Vet. Res. 2017, 13, 378. [Google Scholar] [CrossRef]
  174. Burow, E.; Grobbel, M.; Tenhagen, B.A.; Simoneit, C.; Ladwig, M.; Szabo, I.; Wendt, D.; Banneke, S.; Kasbohrer, A. Antimicrobial susceptibility in faecal Escherichia coli from pigs after enrofloxacin administration in an experimental environment. Berl. Munch. Tierarztl. Wochenschr. 2018, 131, 170–181. [Google Scholar]
  175. Gellin, G.; Langlois, B.; Dawson, K.; Aaron, D. Antibiotic resistance of gram-negative enteric bacteria from pigs in three herds with different histories of antibiotic exposure. Appl. Environ. Microbiol. 1989, 55, 2287–2292. [Google Scholar] [CrossRef] [PubMed]
  176. Mathew, A.G.; Upchurch, W.G.; Chattin, S.E. Incidence of antibiotic resistance in fecal Escherichia coli isolated from commercial swine farms. J. Anim. Sci. 1998, 76, 429. [Google Scholar] [CrossRef]
  177. Mencía-Ares, O.; Argüello, H.; Puente, H.; Gómez-García, M.; Manzanilla, E.G.; Álvarez-Ordóñez, A.; Carvajal, A.; Rubio, P. Antimicrobial resistance in commensal Escherichia coli and Enterococcus spp. is influenced by production system, antimicrobial use, and biosecurity measures on Spanish pig farms. Porc. Health Manag. 2021, 7, 27. [Google Scholar] [CrossRef]
  178. Hickman, R.A.; Leangapichart, T.; Lunha, K.; Jiwakanon, J.; Angkititrakul, S.; Magnusson, U.; Sunde, M.; Järhult, J.D. Exploring the Antibiotic Resistance Burden in Livestock, Livestock Handlers and Their Non-Livestock Handling Contacts: A One Health Perspective. Front. Microbiol. 2021, 12, 651461. [Google Scholar] [CrossRef]
  179. Dewulf, J.; Catry, B.; Timmerman, T.; Opsomer, G.; de Kruif, A.; Maes, D. Tetracycline-resistance in lactose-positive enteric coliforms originating from Belgian fattening pigs: Degree of resistance, multiple resistance and risk factors. Prev. Vet. Med. 2007, 78, 339–351. [Google Scholar] [CrossRef]
  180. Vieira, A.R.; Houe, H.; Wegener, H.C.; Wong, D.M.L.F.; Emborg, H.D.D. Association between tetracycline consumption and tetracycline resistance in Escherichia coli from healthy Danish slaughter pigs. Foodborne Pathog. Dis. 2009, 6, 99–109. [Google Scholar] [CrossRef]
  181. Dunlop, R.H.; McEwen, S.A.; Meek, A.H.; Clarke, R.C.; Black, W.D.; Friendship, R.M. Associations among antimicrobial drug treatments and antimicrobial resistance of fecal Escherichia coli of swine on 34 farrow-to-finish farms in Ontario, Canada. Prev. Vet. Med. 1998, 34, 283–305. [Google Scholar] [PubMed]
  182. Rosengren, L.B.; Waldner, C.L.; Reid-Smith, R.J.; Dowling, P.M.; Harding, J.C. Associations Between Feed and Water Antimicrobial Use in Farrow-to-Finish Swine Herds and Antimicrobial Resistance of Fecal Escherichia coli from Grow-Finish Pigs. Microb. Drug Resist. 2007, 13, 261–270. [Google Scholar] [PubMed]
  183. Harada, K.; Asai, T.; Ozawa, M.; Kojima, A.; Takahashi, T. Farm-Level Impact of Therapeutic Antimicrobial Use on Antimicrobial-Resistant Populations of Escherichia coli Isolates from Pigs. Microb. Drug Resist. 2008, 14, 239–244. [Google Scholar] [PubMed]
  184. Alali, W.Q.; Scott, H.M.; Christian, K.L.; Fajt, V.R.; Harvey, R.B.; Lawhorn, D.B. Relationship between level of antibiotic use and resistance among Escherichia coli isolates from integrated multi-site cohorts of humans and swine. Prev. Vet. Med. 2009, 90, 160–167. [Google Scholar] [CrossRef]
  185. Varga, C.; Rajić, A.; McFall, M.E.; Reid-Smith, R.J.; Deckert, A.E.; Checkley, S.L.; McEwen, S.A. Associations between reported on-farm anti-microbial use practices and observed antimicrobial resistance in generic fecal Escherichia coli isolated from Alberta finishing swine farms. Prev. Vet. Med. 2009, 88, 185–192. [Google Scholar] [CrossRef]
  186. Makita, K.; Goto, M.; Ozawa, M.; Kawanishi, M.; Koike, R.; Asai, T.; Tamura, Y. Multivariable Analysis of the Association Between Antimicrobial Use and Antimicrobial Resistance in Escherichia coli Isolated from Apparently Healthy Pigs in Japan. Microb. Drug Resist. 2016, 22, 28–39. [Google Scholar] [CrossRef]
  187. Akwar, H.; Poppe, C.; Wilson, J.; Reid-Smith, R.J.; Dyck, M.; Waddington, J.; Shang, D.; McEwen, S.A. Associations of antimicrobial uses with antimicrobial resistance of fecal Escherichia coli from pigs on 47 farrow-to-finish farms in Ontario and British Columbia. Can. J. Vet. Res. 2008, 72, 202–210. [Google Scholar]
  188. European Medicines Agency (EMA). Categorisation of Antibiotics for Use in Animals for Prudent and Responsible Use. 2019. Available online: https://www.ema.europa.eu/en/documents/report/categorisation-antibiotics-european-union-answer-request-european-commission-updating-scientific_en.pdf (accessed on 6 June 2023).
  189. Regulation (EU) 2019/6 of the European Parliament and of the Council of 11 December 2018 on Veterinary Medicinal Products and Repealing Directive 2001/82/EC (Text with EEA relevance). Off. J. Eur. Union 2019, L4, 43–167. Available online: http://data.europa.eu/eli/reg/2019/6/oj (accessed on 6 July 2021).
  190. Lambrecht, E.; Meervenne, E.; Boon, N.; de Wiele, T.; Wattiau, P.; Herman, L.; Heyndrickx, M.; Coillie, E. Characterization of Cefotaxime- and Ciprofloxacin-Resistant Commensal Escherichia coli Originating from Belgian Farm Animals Indicates High Antibiotic Resistance Transfer Rates. Microb. Drug Resist. 2018, 24, 707–717. [Google Scholar] [CrossRef]
  191. Jacoby, G.A. AmpC β-Lactamases. Clin. Microbiol. Rev. 2009, 22, 161–182. [Google Scholar] [CrossRef] [PubMed]
  192. Bush, K.; Jacoby, G.A. Updated Functional Classification of β-Lactamases. Antimicrob. Agents Chemother. 2010, 54, 969–976. [Google Scholar] [CrossRef]
  193. Bush, K.; Bradford, P.A. β-Lactams and β-Lactamase Inhibitors: An Overview. Cold Spring Harb. Perspect. Med. 2016, 6, a025247. [Google Scholar] [CrossRef] [PubMed]
  194. Livermore, D.M.; Canton, R.; Gniadkowski, M.; Nordmann, P.; Rossolini, G.; Arlet, G.; Ayala, J.; Coque, T.M.; Kern-Zdanowicz, I.; Luzzaro, F.; et al. CTX-M: Changing the face of ESBLs in Europe. J. Antimicrob. Chemother. 2007, 59, 165–174. [Google Scholar] [CrossRef] [PubMed]
  195. Woerther, P.L.; Burdet, C.; Chachaty, E.; Andremont, A. Trends in Human Fecal Carriage of Extended-Spectrum β-Lactamases in the Community: Toward the Globalization of CTX-M. Clin. Microbiol. Rev. 2013, 26, 744–758. [Google Scholar] [CrossRef] [PubMed]
  196. Bevan, E.R.; Jones, A.M.; Hawkey, P.M. Global epidemiology of CTX-M β-lactamases: Temporal and geographical shifts in genotype. J. Antimicrob. Chemoth. 2017, 72, 2145–2155. [Google Scholar] [CrossRef]
  197. Liu, J.H.; Wei, S.Y.; Ma, J.Y.; Zeng, Z.L.; Lü, D.H.; Yang, G.X.; Chen, Z.L. Detection and characterisation of CTX-M and CMY-2 β-lactamases among Escherichia coli isolates from farm animals in Guangdong Province of China. Int. J. Antimicrob. Agents 2007, 29, 576–581. [Google Scholar] [CrossRef]
  198. Tian, G.B.; Wang, H.N.; Zou, L.K.; Tang, J.N.; Zhao, Y.W.; Ye, M.Y.; Tang, J.Y.; Zhang, Y.; Zhang, A.Y.; Yang, X.; et al. Detection of CTX-M-15, CTX-M-22, and SHV-2 Extended-Spectrum β-Lactamases (ESBLs) in Escherichia coli Fecal-Sample Isolates from Pig Farms in China. Foodborne Pathog. Dis. 2009, 6, 297–304. [Google Scholar] [CrossRef]
  199. Zheng, H.; Zeng, Z.; Chen, S.; Liu, Y.; Yao, Q.; Deng, Y.; Chen, X.; Lv, L.; Zhuo, C.; Chen, Z.; et al. Prevalence and characterisation of CTX-M β-lactamases amongst Escherichia coli isolates from healthy food animals in China. Int. J. Antimicrob. Agents 2012, 39, 305–310. [Google Scholar] [CrossRef]
  200. Tamang, M.; Nam, H.M.; Kim, S.R.; Chae, M.; Jang, G.C.; Jung, S.C.; Lim, S.K. Prevalence and Molecular Characterization of CTX-M β-Lactamase-Producing Escherichia coli Isolated from Healthy Swine and Cattle. Foodborne Pathog. Dis. 2013, 10, 13–20. [Google Scholar] [CrossRef]
  201. Rao, L.; Lv, L.; Zeng, Z.; Chen, S.; He, D.; Chen, X.; Wu, C.; Wang, Y.; Yang, T.; Wu, P.; et al. Increasing prevalence of extended-spectrum cephalosporin-resistant Escherichia coli in food animals and the diversity of CTX-M genotypes during 2003–2012. Vet. Microbiol. 2014, 172, 534–541. [Google Scholar] [CrossRef] [PubMed]
  202. Xu, G.; An, W.; Wang, H.; Zhang, X. Prevalence and characteristics of extended-spectrum β-lactamase genes in Escherichia coli isolated from piglets with post-weaning diarrhea in Heilongjiang province, China. Front. Microbiol. 2015, 6, 1103. [Google Scholar] [CrossRef]
  203. Song, H.J.; Moon, D.C.; Kim, S.J.; Mechesso, A.F.; Choi, J.H.; Boby, N.; Kang, H.Y.; Na, S.H.; Yoon, S.S.; Lim, S.K. Antimicrobial Resistance Profiles and Molecular Characteristics of Extended-Spectrum β-Lactamase-Producing Escherichia coli Isolated from Healthy Cattle and Pigs in Ko-rea. Foodborne Pathog. Dis. 2023, 20, 7–16. [Google Scholar] [CrossRef] [PubMed]
  204. Lee, S.; An, J.U.; Guk, J.H.; Song, H.; Yi, S.; Kim, W.H.; Cho, S. Prevalence, Characteristics and Clonal Distribution of Extended-Spectrum β-Lactamase- and AmpC β-Lactamase-Producing Escherichia coli Following the Swine Production Stages, and Potential Risks to Humans. Front. Microbiol. 2021, 12, 710747. [Google Scholar] [CrossRef] [PubMed]
  205. Song, J.; Oh, S.S.; Kim, J.; Park, S.; Shin, J. Clinically Relevant Extended-Spectrum β-Lactamase-Producing Escherichia coli Isolates From Food Animals in South Korea. Front. Microbiol. 2020, 11, 604. [Google Scholar] [CrossRef]
  206. Seo, K.W.; Do, K.H.; Jung, C.M.; Lee, S.W.; Lee, Y.J.; Lim, S.K.; Lee, W.K. Comparative genetic characterisation of third-generation cephalosporin-resistant Escherichia coli isolated from integrated and conventional pig farm in Korea. J. Glob. Antimicrob. Resist. 2023, 34, 74–82. [Google Scholar] [CrossRef]
  207. Frye, J.G.; Jackson, C.R. Genetic mechanisms of antimicrobial resistance identified in Salmonella enterica, Escherichia coli, and Enterococcus spp. isolated from U.S. food animals. Front. Microbiol. 2013, 4, 135. [Google Scholar] [CrossRef]
  208. Jahanbakhsh, S.; Smith, M.G.; Kohan-Ghadr, H.R.; Letellier, A.; Abraham, S.; Trott, D.J.; Fairbrother, J. Dynamics of extended-spectrum cephalosporin resistance in pathogenic Escherichia coli isolated from diseased pigs in Quebec, Canada. Int. J. Antimicrob. Agents 2016, 48, 194–202. [Google Scholar] [CrossRef]
  209. Hayer, S.; Lim, S.; Hong, S.; Elnekave, E.; Johnson, T.; Rovira, A.; Vannucci, F.; Clayton, J.B.; Perez, A.; Alvarez, J. Genetic Determinants of Resistance to Extended-Spectrum Cephalosporin and Fluoroquinolone in Escherichia coli Isolated from Diseased Pigs in the United States. Msphere 2020, 5, 10–128. [Google Scholar] [CrossRef]
  210. European Food Safety Authority (EFSA) and European Centre for Disease Prevention and Control (ECDC), 2021. The European Union Summary Report on Antimicrobial Resistance in zoonotic and indicator bacteria from humans, animals and food in 2018/2019. EFSA J. 2021, 19, 6490. [Google Scholar] [CrossRef]
  211. Ewers, C.; de Jong, A.; Prenger-Berninghoff, E.; Garch, F.; Leidner, U.; Tiwari, S.K.; Semmler, T. Genomic Diversity and Virulence Potential of ESBL- and AmpC-β-Lactamase-Producing Escherichia coli Strains From Healthy Food Animals Across Europe. Front. Microbiol. 2021, 12, 626774. [Google Scholar] [CrossRef]
  212. Endimiani, A.; Rossano, A.; Kunz, D.; Overesch, G.; Perreten, V. First countrywide survey of third-generation cephalosporin-resistant Escherichia coli from broilers, swine, and cattle in Switzerland. Diagn. Micr. Infect. Dis. 2012, 73, 31–38. [Google Scholar] [CrossRef]
  213. Wu, G.; Day, M.J.; Mafura, M.T.; Nunez-Garcia, J.; Fenner, J.J.; Sharma, M.; van Essen-Zandbergen, A.; Rodríguez, I.; Dierikx, C.; Kadlec, K.; et al. Comparative Analysis of ESBL-Positive Escherichia coli Isolates from Animals and Humans from the UK, The Netherlands and Germany. PLoS ONE 2013, 8, e75392. [Google Scholar] [CrossRef] [PubMed]
  214. Valentin, L.; Sharp, H.; Hille, K.; Seibt, U.; Fischer, J.; Pfeifer, Y.; Michael, G.B.; Nickel, S.; Schmiedel, J.; Falgenhauer, L.; et al. Subgrouping of ESBL-producing Escherichia coli from animal and human sources: An approach to quantify the distribution of ESBL types between different reservoirs. Int. J. Med. Microbiol. 2014, 304, 805–816. [Google Scholar] [CrossRef] [PubMed]
  215. García-Cobos, S.; Köck, R.; Mellmann, A.; Frenzel, J.; Friedrich, A.W.; Rossen, J.W. Molecular Typing of Enterobacteriaceae from Pig Holdings in North-Western Germany Reveals Extended-Spectrum and AmpC β-Lactamases Producing but no Carbapenem Resistant Ones. PLoS ONE 2015, 10, e0134533. [Google Scholar] [CrossRef] [PubMed]
  216. Lalak, A.; Wasyl, D.; Zając, M.; Skarżyńska, M.; Hoszowski, A.; Samcik, I.; Woźniakowski, G.; Szulowski, K. Mechanisms of cephalosporin resistance in indicator Escherichia coli isolated from food animals. Vet. Microbiol. 2016, 194, 69–73. [Google Scholar] [CrossRef]
  217. Rodrigues, C.; Machado, E.; Peixe, L.; Novais, Â. IncI1/ST3 and IncN/ST1 plasmids drive the spread of blaTEM-52 and blaCTX-M-1/-32 in diverse Escherichia coli clones from different piggeries. J. Antimicrob. Chemoth. 2013, 68, 2245–2248. [Google Scholar] [CrossRef]
  218. Fournier, C.; Aires-de-Sousa, M.; Nordmann, P.; Poirel, L. Occurrence of CTX-M-15- and MCR-1-producing Enterobacteales in pigs in Portugal: Evidence of direct links with antibiotic selective pressure. Int. J. Antimicrob. Agents 2020, 55, 105802. [Google Scholar] [CrossRef]
  219. Zelendova, M.; Dolejska, M.; Masarikova, M.; Jamborova, I.; Vasek, J.; Smola, J.; Manga, I.; Cizek, A. CTX-M-producing Escherichia coli in pigs from a Czech farm during production cycle. Lett. Appl. Microbiol. 2020, 71, 369–376. [Google Scholar] [CrossRef] [PubMed]
  220. Hansen, K.; Bortolaia, V.; Damborg, P.; Guardabassi, L. Strain Diversity of CTX-M-Producing Enterobacteriaceae in Individual Pigs: In-sights into the Dynamics of Shedding during the Production Cycle. Appl. Environ. Microbiol. 2014, 80, 6620–6626. [Google Scholar] [CrossRef]
  221. Abraham, S.; Kirkwood, R.N.; Laird, T.; Saputra, S.; Mitchell, T.; Singh, M.; Linn, B.; Abraham, R.J.; Pang, S.; Gordon, D.M.; et al. Dissemination and persistence of extended-spectrum cephalosporin-resistance encoding IncI1-blaCTXM-1 plasmid among Escherichia coli in pigs. ISME J. 2018, 12, 2352–2362. [Google Scholar] [CrossRef]
  222. Moor, J.; Aebi, S.; Rickli, S.; Mostacci, N.; Overesch, G.; Oppliger, A.; Hilty, M. Dynamics of extended-spectrum cephalosporin-resistant E. coli in pig farms: A longitudinal study. Int. J. Antimicrob. Agents 2021, 58, 106382. [Google Scholar] [CrossRef]
  223. Dohmen, W.; Dorado-García, A.; Bonten, M.J.M.; Wagenaar, J.A.; Mevius, D.; Heederik, D.J.J. Risk factors for ESBL-producing Escherichia coli on pig farms: A longitudinal study in the context of reduced use of antimicrobials. PLoS ONE 2017, 12, e0174094. [Google Scholar] [CrossRef] [PubMed]
  224. von Salviati, C.; Friese, A.; Roschanski, N.; Laube, H.; Guerra, B.; Käsbohrer, A.; Kreienbrock, L.; Roesler, U. Extended-spectrum beta-lactamases (ESBL)/AmpC beta-lactamases-producing Escherichia coli in German fattening pig farms: A longitudinal study. Berl. Und Münchener Tierärztliche Wochenschr. 2014, 127, 412–419. [Google Scholar]
  225. Poulin-Laprade, D.; Brouard, J.S.; Gagnon, N.; Turcotte, A.; Langlois, A.; Matte, J.J.; Carrillo, C.D.; Zaheer, R.; McAllister, T.A.; Topp, E.; et al. Resistance Determinants and Their Genetic Context in Enterobacteria from a Longitudinal Study of Pigs Reared under Various Husbandry Conditions. Appl. Environ. Microbiol. 2021, 87, e02612-20. [Google Scholar] [CrossRef]
  226. Nguyen, N.T.; Nguyen, H.M.; Nguyen, C.V.; Nguyen, T.V.; Nguyen, M.T.; Thai, H.Q.; Ho, M.H.; Thwaites, G.; Ngo, H.T.; Baker, S.; et al. Use of Colistin and Other Critical Antimicrobials on Pig and Chicken Farms in Southern Vietnam and Its Association with Resistance in Commensal Escherichia coli Bacteria. Appl. Environ. Microbiol. 2016, 82, 3727–3735. [Google Scholar] [CrossRef]
  227. Jørgensen, C.J.; Cavaco, L.M.; Hasman, H.; Emborg, H.D.; Guardabassi, L. Occurrence of CTX-M-1-producing Escherichia coli in pigs treated with ceftiofur. J. Antimicrob. Chemother. 2007, 59, 1040–1042. [Google Scholar] [CrossRef] [PubMed]
  228. Lutz, E.A.; McCarty, M.J.; Mollenkopf, D.F.; Funk, J.A.; Gebreyes, W.A.; Wittum, T.E. Ceftiofur Use in Finishing Swine Barns and the Recovery of Fecal Escherichia coli or Salmonella spp. Resistant to Ceftriaxone. Foodborne Pathog. Dis. 2011, 8, 1229–1234. [Google Scholar] [CrossRef] [PubMed]
  229. Andersen, V.D.; Jensen, V.F.; Vigre, H.; Andreasen, M.; Agersø, Y. The use of third and fourth generation cephalosporins affects the occurrence of extended-spectrum cephalosporinase-producing Escherichia coli in Danish pig herds. Vet. J. 2015, 204, 345–350. [Google Scholar] [CrossRef]
  230. Agersø, Y.; Aarestrup, F.M. Voluntary ban on cephalosporin use in Danish pig production has effectively reduced extended-spectrum cephalosporinase-producing Escherichia coli in slaughter pigs. J. Antimicrob. Chemoth. 2013, 68, 569–572. [Google Scholar] [CrossRef] [PubMed]
  231. Jacoby, G.A. Mechanisms of resistance to quinolones. Clin. Infect. Dis. 2005, 41, S120–S126. [Google Scholar] [CrossRef]
  232. Strahilevitz, J.; Jacoby, G.A.; Hooper, D.C.; Robicsek, A. Plasmid-Mediated Quinolone Resistance: A Multifaceted Threat. Clin. Microbiol. Rev. 2009, 22, 664–689. [Google Scholar] [CrossRef]
  233. Taylor, N.M.; Davies, R.H.; Ridley, A.; Clouting, C.; Wales, A.D.; Clifton-Hadley, F.A. A survey of fluoroquinolone resistance in Escherichia coli and thermophilic Campylobacter spp. on poultry and pig farms in Great Britain. J. Appl. Microbiol. 2008, 105, 1421–1431. [Google Scholar] [PubMed]
  234. Rhouma, M.; Madec, J.Y.; Laxminarayan, R. Colistin: From the shadows to a One Health approach for addressing antimicrobial resistance. Int. J. Antimicrob. Agents 2023, 61, 106713. [Google Scholar] [CrossRef]
  235. Liu, Y.Y.; Wang, Y.; Walsh, T.R.; Yi, L.X.; Zhang, R.; Spencer, J.; Doi, Y.; Tian, G.; Dong, B.; Huang, X.; et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: A microbiological and molecular biological study. Lancet Infect. Dis. 2016, 16, 161–168. [Google Scholar] [CrossRef]
  236. Zhang, S.; Abbas, M.; Rehman, M.; Wang, M.; Jia, R.; Chen, S.; Liu, M.; Zhu, D.; Zhao, X.; Gao, Q.; et al. Updates on the global dissemination of colistin-resistant Escherichia coli: An emerging threat to public health. Sci. Total Environ. 2021, 799, 149280. [Google Scholar] [CrossRef] [PubMed]
  237. Lee, S.; An, J.U.; Woo, J.; Song, H.; Yi, S.; Kim, W.H.; Lee, J.H.; Ryu, S.; Cho, S. Prevalence, Characteristics, and Clonal Distribution of Escherichia coli Carrying Mobilized Colistin Resistance Gene mcr-1.1 in Swine Farms and Their Differences According to Swine Production Stages. Front. Microbiol. 2022, 13, 873856. [Google Scholar] [CrossRef] [PubMed]
  238. Food and Agriculture Organization (FAO). The FAO Action Plan on Antimicrobial Resistance 2021–2025; FAO: Rome, Italy, 2021. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of potential transmission pathways of AMR Escherichia coli and/or their associated ARGs between pigs, humans, and the environment. The smaller circular icon associated with the human and environment ecosystems represents AMR transmission within the microbiota of the relevant ecosystem. Legend: AMR—antimicrobial resistance; ARG—antimicrobial resistance gene; MGE—mobile genetic element.
Figure 1. Schematic representation of potential transmission pathways of AMR Escherichia coli and/or their associated ARGs between pigs, humans, and the environment. The smaller circular icon associated with the human and environment ecosystems represents AMR transmission within the microbiota of the relevant ecosystem. Legend: AMR—antimicrobial resistance; ARG—antimicrobial resistance gene; MGE—mobile genetic element.
Antibiotics 12 01616 g001
Table 1. Summary of antimicrobial resistance in commensal Escherichia coli of porcine origin extracted from the most recent data of monitoring programmes in the EU/EEA, UK, USA, Canada, and Japan. Data from selected European countries participating in the EFSA and ECDC monitoring programme are included.
Table 1. Summary of antimicrobial resistance in commensal Escherichia coli of porcine origin extracted from the most recent data of monitoring programmes in the EU/EEA, UK, USA, Canada, and Japan. Data from selected European countries participating in the EFSA and ECDC monitoring programme are included.
Antimicrobial a
CountryYear bTETSUL cTMPSXTAMPCHLSTRGENAXO dCTX dCIP dCIP HL dAZM dCOL dCSMDR
Denmark202129.2%41.5%30.8%-38.5%4.6%-0.0%-1.5%0.0%-4.6%0.0%52.3%33.8%
France202142.2%29.3%21.1%-25.4%8.6%-0.0%-0.9%2.6%0.4%1.3%0.0%44.0%23.7%
Germany202132.1%29.5%23.7%-28.9%6.3%-2.6%-0.0%1.6%0.5%2.6%0.0%49.5%22.6%
Ireland202151.8%37.1%36.5%-28.2%9.4%-3.5%-0.0%2.4%0.0%0.6%0.0%38.2%32.9%
Netherlands202131.0%24.0%24.3%-22.3%9.3%-0.7%-0.0%2.0%0.0%1.7%0.0%50.7%20.3%
Spain202178.8%58.8%60.0%-83.5%41.2%-4.7%-1.2%50.6%11.8%4.7%0.0%6.5%78.8%
Sweden202116.8%22.5%19.7%-24.9%7.5%-0.0%-0.6%1.7%0.0%0.6%0.0%63.6%19.7%
EU/EEA e202145.9%33.8%25.9%-32.8%11.8%-1.1%-0.9%6.4%1.2%1.6%0.0%38.3%31.2%
UK f202152.7%40.5%37.6%-33.3%18.6%-2.1%-1.3%4.6%--0.0%--
USA g202166.5%20.3%-9.7%25.0%7.2%-3.8%9.3%-10.2%3.0%0.4%-27.1%16.1%
Canada h201955.5%35.1%-13.1%29.9%12.4%40.9%0.0%2.2%--0.0%0.0%-25.5%-
Japan i201755.4%--26.5%33.7%21.7%41.0%3.6%-1.2%---0.0%--
a Antimicrobials: TET—tetracycline; SUL—sulfamethoxazole or sulfisoxazole (see c); TMP—trimethoprim; SXT—trimethoprim/sulfamethoxazole; AMP—ampicillin; CHL—chloramphenicol; STR—streptomycin; GEN—gentamicin; AXO—ceftriaxone; CTX—cefotaxime; CIP—ciprofloxacin (MIC > 0.06 mg/L); CIP HL—ciprofloxacin (MIC > 1 mg/L); AZM—azithromycin; COL—colistin; CS—complete susceptibility to all antimicrobials tested; MDR—multidrug resistance, resistance to antimicrobials in three or more classes. b Year of sampling. c Sulfamethoxazole is the sulphonamide representative in the EFSA and ECDC testing panel. Sulfisoxazole is the sulphonamide representative in the NARMS and CIPARS panels. d Highest Priority Critically Important Antimicrobial [8] e Participating EU/EEA countries submit data every two years to the EFSA and ECDC monitoring programme. Data from seven selected countries of interest are shown. The overall EU/EEA data, highlighted in bold, represents the median for all 32 participating countries. Antimicrobial susceptibility interpreted according to ECOFFs defined by EUCAST [33]. f United Kingdom Veterinary Antimicrobial Resistance and Sales Surveillance 2021 (UK-VARSS). Antimicrobial susceptibility interpreted according to ECOFFs defined by EUCAST [31]. g The National Antimicrobial Resistance Monitoring System (NARMS). Antimicrobial susceptibility interpreted according to CLSI M100-Ed30 [68]. h Canadian Integrated Program for Antimicrobial Resistance Surveillance (CIPARS). Antimicrobial susceptibility interpreted according to CLSI M100-S26 [87]. i Japanese Veterinary Antimicrobial Resistance Monitoring System (JVARM). Antimicrobial susceptibility interpreted according to CLSI M100-S27 [88]. Legend: CLSI—Clinical and Laboratories Standards Institute; ECOFF—Epidemiologic cut off value; EEA—European Economic Area; EU—European Union; EUCAST—European Committee on Antimicrobial Susceptibility Testing; UK—United Kingdom; USA—United States of America.
Table 2. Summary of resistance to selected antimicrobials in commensal Escherichia coli from healthy pigs at or before slaughter, extracted from selected published studies.
Table 2. Summary of resistance to selected antimicrobials in commensal Escherichia coli from healthy pigs at or before slaughter, extracted from selected published studies.
StudyAntimicrobial a
CountryYear bTETSULTMPSXTAMPSTRGENCHLCTXTIOAXOCIP
Europe
Spain [89]200095.6%87.8%83.4%-72.2%-8.0%59.5%----
Spain [90]200168.0%--48.0%29%-7.0%15.0%0%--3.0%
Portugal [63]201393.9%--69.7%68.2%77.3%4.5%36.4%0%--1.5%
Poland [64]201348.9%35.8%16.3%-29.5%42.6%2.6%18.9%2.6%--6.3%
Ireland [65]201659.0%--27.6%18.0%33.3%5.8%9.6%0%0%-0%
Estonia [67]201932.5%30.0%22.4%-58.7%39.2%12.5%5.8%2.5%-0%5.8%
Denmark c [66]201642.3%24.6%23.1%-25.0%44.2%5.8%0%0%--0%
France c [66]201674.5%--40.4%14.9%66.0%7.5%17.0%0%--4.3%
Italy c [66]201674.4%61.6%50.4%-62.4%61.6%6.4%30.4%0%--12.0%
Sweden c [66]201614.1%25.4%19.7%-18.3%25.4%1.4%1.4%0%--1.4%
Denmark [91]201236.0%--14.7%24.0%-0%6.7%0%--0%
France [91]201283.2%--43.6%24.8%-2.0%20.8%0%--0%
Germany [91]201264.4%--33.7%33.7%-0%13.5%0%--1.0%
Netherlands [91]201267.9%--42.1%25.7%-0%14.3%0%--0%
Spain [91]201294.0%--66.0%66.0%-5.0%42.0%0%--0%
Europe d [92]202253.3%--29.5%35.3%-2.2%21.3%0.8%--1.6%
France d [92]202261.7%--29.0%29.4%-0.5%13.6%0.5%--0.9%
Germany d [92]202232.9%--18.6%27.1%-0.5%6.2%2.9%--1.0%
Netherlands d [92]202243.1%--24.1%24.5%-0.5%31.0%0.0%--0.5%
Spain d [92]202281.6%--50.2%66.7%-5.5%39.8%0.5%--6.0%
UK d [92]202248.5%--26.5%30.4%-4.4%16.7%0.0%--0.0%
North America
Canada [93]199871.3%38.2%--29.1%-0.6%-----
Canada [94]200878.9%49.9%-6.4%30.6%49.6%1.1%17.6%-0%0%0%
Canada [95]200866.8%46.0%-7.4%18.6%33.4%0.8%17.3%-0.1%0%0%
Canada [96]200873.0%46.6%-2.6%25.7%27.5%0.4%15.5%-0%0.43%0%
USA c [97]200790.9%31.6%-1.9%24.1%28.9%0.8%8.2%-0.3%2.0%0%
Australia
Australia [98]2016----29.4%-17.5%--1.8%--
Australia [99]201867.7%--34.3%60.2%33.9%0%22.4%-0%0%1.0%
Asia
Korea [100]200796.3%--38.8%66.1%66.8%42.0%47.6%1.0%--7.8%
Korea d [101]201489.9%---71.3%61.2%23.2%68.4%---8.4%
Korea d [102]202273.9%--84.8%79.4%74.5%17.6%80.0%-5.5%-14.5%
China [74]201998.3%--71.6%90.0%-21.7%75.0%---21.7%
China [75]202073.5%--71.6%58.0%53.0%21.6%-16.7%--23.9%
Africa
Tanzania [72]201572.9%--60.0%38.6%50.0%--24.3%--10%
Tanzania [73]202151.3%--47.7%46.4%-26.0%27.3%29.5%--28.6%
Nigeria [69]201550.0%--17.9%10.7%-0%-3.6%-3.6%3.6%
Rwanda [71]202126.7%---12.6%13.3%-2.2%0.7%-0.7%0%
Uganda [70]202153.9%88.5%17.3%-11.5%-3.8%5.7%7.7%--7.6%
a Antimicrobial: TET—tetracycline; SUL—sulfamethoxazole or sulfisoxazole; TMP—trimethoprim; SXT—trimethoprim/sulfamethoxazole; AMP -ampicillin; STR—streptomycin; GEN—gentamicin; CHL—chloramphenicol; AXO—ceftriaxone; TIO—ceftiofur; CTX—cefotaxime; CIP—ciprofloxacin, b Year of publication c Study compared conventional and organic/antibiotic free systems. Results from conventional farms only are shown, d Multi-year study: results from final year of study are shown. Legend: UK—United Kingdom; USA—United States of America.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

O’Neill, L.; Manzanilla, E.G.; Ekhlas, D.; Leonard, F.C. Antimicrobial Resistance in Commensal Escherichia coli of the Porcine Gastrointestinal Tract. Antibiotics 2023, 12, 1616. https://doi.org/10.3390/antibiotics12111616

AMA Style

O’Neill L, Manzanilla EG, Ekhlas D, Leonard FC. Antimicrobial Resistance in Commensal Escherichia coli of the Porcine Gastrointestinal Tract. Antibiotics. 2023; 12(11):1616. https://doi.org/10.3390/antibiotics12111616

Chicago/Turabian Style

O’Neill, Lorcan, Edgar García Manzanilla, Daniel Ekhlas, and Finola C. Leonard. 2023. "Antimicrobial Resistance in Commensal Escherichia coli of the Porcine Gastrointestinal Tract" Antibiotics 12, no. 11: 1616. https://doi.org/10.3390/antibiotics12111616

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