Next Article in Journal
The Effect of Bifidobacterium animalis subsp. lactis Bl-04 on Influenza A Virus Infection in Mice
Previous Article in Journal
The Effects of Lactobacillus johnsonii on Diseases and Its Potential Applications
Previous Article in Special Issue
Drinking Pipes and Nipple Drinkers in Pig Abattoir Lairage Pens—A Source of Zoonotic Pathogens as a Hazard to Meat Safety
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Antimicrobial Resistance in Bacteria from Meat and Meat Products: A One Health Perspective

by
Sara Conceição
1,
Maria Cristina Queiroga
1,2 and
Marta Laranjo
1,2,*
1
MED—Mediterranean Institute for Agriculture, Environment and Development & CHANGE—Global Change and Sustainability Institute, Institute for Advanced Studies and Research, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
2
Departamento de Medicina Veterinária, Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(10), 2581; https://doi.org/10.3390/microorganisms11102581
Submission received: 25 July 2023 / Revised: 13 October 2023 / Accepted: 14 October 2023 / Published: 17 October 2023

Abstract

:
According to the 2030 Agenda of the United Nations, one of the sustainable development goals is to ensure sustainable consumption and production patterns. The need to ensure food safety includes, other than microbiological hazards, concerns with antimicrobial-resistant (AMR) bacteria. The emergence of resistant bacteria in the food industry is essentially due to the abusive, and sometimes incorrect, administration of antimicrobials. Although not allowed in Europe, antimicrobials are often administered to promote animal growth. Each time antimicrobials are used, a selective pressure is applied to AMR bacteria. Moreover, AMR genes can be transmitted to humans through the consumption of meat-harbouring-resistant bacteria, which highlights the One Health dimension of antimicrobial resistance. Furthermore, the appropriate use of antimicrobials to ensure efficacy and the best possible outcome for the treatment of infections is regulated through the recommendations of antimicrobial stewardship. The present manuscript aims to give the current state of the art about the transmission of AMR bacteria, particularly methicillin-resistant S. aureus, ESBL-producing Enterobacteriaceae, and vancomycin-resistant Enterococcus spp., along with other ESKAPE bacteria, from animals to humans through the consumption of meat and meat products, with emphasis on pork meat and pork meat products, which are considered the most consumed worldwide.

1. Introduction

Over the last few decades, an increase in antimicrobial-resistant (AMR) bacteria has been observed, including resistance to antimicrobials not authorized in veterinary medicine, which have been reported in meat products [1]. More recently, several policies have been designed to reduce AMR. New therapeutic strategies have been developed, such as the use of bacteriophages, antimicrobial peptides, and phytobiotics such as essential oils or propolis ethanol extracts [2,3,4,5,6,7,8,9]. In addition, the “Antimicrobial Stewardship” strategy was developed in 2007 to obtain better clinical outcomes for the treatment of infections involving a careful selection of antimicrobials, as well as their route, dose, and treatment duration [10]. Furthermore, One Health aims to achieve optimal human health and well-being while also being internally related to animal health and the environment. One Health promotes the fight against antimicrobial resistance because both humans and animals are affected by the same bacteria and are also treated with the same antimicrobials. The One Health approach is considered a collaboration between different sectors, developed in 2003 by the now joint quadripartite consortium, including the Food and Agriculture Organisation of the United Nations (FAO), the World Health Organisation (WHO), the World Organisation for Animal Health (WOAH, founded as OIE), and the United Nations Environment Programme (UNEP) [11]. Moreover, in September 2015, the United Nations developed a global action plan with 193 countries. This action plan, named “Transforming Our World: The 2030 Agenda for Sustainable Development”, has sustainable development as its main goal and has 17 Sustainable Development Goals (SDGs) [12,13,14]. For these SDGs to be fulfilled, around 170 targets were established to address several issues. Examples of these issues are climate change, environmental degradation, and social inequality [14]. However, SDGs are also related to food, namely SDG 12, which aims to promote responsible and sustainable food production and consumption [15]. Thus, SGD 12 is directly linked to one of the current problems regarding food, the antimicrobial-resistant bacteria present in food.
Antimicrobial resistance is considered a complex problem and a global health concern for both humans and animals. Around 2.8 million human cases of infections by antimicrobial-resistant bacteria and 700,000 deaths are reported annually, and this number could reach 10 million by 2050 if AMR is not reduced [16,17]. Antimicrobials have been used extensively and without respecting the therapy guidelines, mostly in low- and middle-income countries [18]. Resistant bacteria have been found in humans, animals, food, and the environment, leading to the transmission of resistance genes across bacterial species and between animals, humans, and the environment [18,19,20]. Bacteria isolated from food-producing animals have shown high AMR for most antimicrobials used in human medicine. About 54% of Escherichia coli and Klebsiella spp. showed high resistance to third-generation cephalosporine [16,18]. The existence of AMR bacteria in foods is mainly due to the excessive use of antimicrobials in food-producing animals and, consequently, the transmission of the AMR bacteria to humans through consumption (also known as “farm-to-fork” transmission) [21,22].
The WHO reported high levels of bacterial AMR worldwide, highlighting the need for a One Health approach to deal with the AMR crisis. The One Health approach works at a local, national, and global level, with the collaboration of policymakers, stakeholders, practitioners, and researchers [18,23].
There are three microbial groups in food products for which AMR can be considered a problem: Staphylococcus spp. (particularly, methicillin-resistant S. aureus), extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae, and some lactic acid bacteria (mainly, vancomycin-resistant Enterococcus spp.) [24,25,26]. Currently, the most threatening multidrug-resistant (MDR) bacteria belong to the ESKAPE group, characterized by the ability to escape the action of various classes of antimicrobials, whether in humans or animals. ESKAPE bacteria comprise Enterococcus (E.) faecium, Staphylococcus (S.) aureus, Klebsiella (K.) pneumoniae, Acinetobacter (A.) baumannii, Pseudomonas (P.) aeruginosa, and Enterobacter (E.) species [2,22,27].
The present review aims to analyse the state of the art related to the AMR in various types of meat and meat products, with an emphasis on pork meat and pork meat products, which are considered the most consumed worldwide. In this type of food, there are three microbiota groups of interest, namely methicillin-resistant S. aureus, ESBL-producing Enterobacteriaceae, and vancomycin-resistant Enterococcus spp. Moreover, ESKAPE bacteria, which include bacteria from the three abovementioned groups, are also addressed as they have the highest impact on AMR bacteria in the food industry [28,29,30,31].

2. The One Health Approach

Antimicrobials have dramatically improved human and animal health. However, the effectiveness of antimicrobials has decreased over the years, giving rise to resistant bacterial strains. Due to the excessive use of antimicrobials in hospitals, in the community, and the livestock sector, there was an emergence of MDR microorganisms. This led to a global AMR increase and a threat to public health as the existence of AMR bacteria hampers the treatment of diseases. Additionally, resistant bacteria may enter the food chain through consumption, increasing the risk of AMR in food pathogens [32,33]. Without effective antimicrobials, healthcare costs, disease occurrence, and mortality rates highly increase. The One Health approach is an integrated and unifying strategy towards the sustainable health of ecosystems, humans, and animals [34,35]. To fight the increase in AMR, the One Health approach establishes communication channels between different sectors for the development and implementation of AMR surveillance programs, achieving optimal health for humans, animals, and ecosystems [19,36].
Additionally, the One Health approach to antimicrobial stewardship is an ecological concept, and its main purpose is to improve prescribing practices by doctors and veterinarians [23,37]. To fulfil the purpose of One Health, there must be a surveillance of AMR transmission elements considered relevant to define the AMR transmission process between humans, animals, plants, and the environment [37].
Specifically concerning the pig industry, the surveillance must focus on the whole value chain, namely the production, slaughtering, and processing steps. Mitigation measures may include hygiene measures throughout the whole chain but also monitoring strategies, promoting both the use of biosafety methods and vaccine research and development [38]. Lately, innate immunomodulation is a new strategy that is currently being studied, where the innate immune memory is achieved through the stimulation of innate immune cells with non-related stimuli [39]. This phenomenon has been recently reported in pigs [40].
One Health, however, is a multi-hierarchical system; there is a problem with predictions, namely how a change in a particular level of the hierarchy affects the remaining levels. Computer science managed to solve this problem with membrane computing modelling, which was recently applied to AMR prediction [37,41]. Through the One Health approach, it will be possible to develop new biochemical, microbiological, ecological, computational, and bioinformatics techniques, which will be necessary to understand and, also, to control the problem of AMR globally [37,42,43].

3. Antimicrobial Activity

Antimicrobials are natural, semi-synthetic, or synthetic compounds capable of killing bacteria or preventing bacterial growth. These are used in the treatment of bacterial infections in humans and animals, or as feed additives or synthetic growth promoters in animals and aquaculture [33].
Antimicrobial activity may be divided into five main mechanisms, which are summarised in Figure 1 [44,45].

3.1. Inhibition of Cell Wall Synthesis

The bacterial cell wall is composed of peptidoglycan that generates mechanical support and allows the bacteria to survive under extreme situations (e.g., osmotic pressure changes) [44,46]. Peptidoglycan is a polymer formed by chains of glycans, formed by disaccharide subunits of N-acetylglucosamine and acetylmuramic acid, cross-linked by pentapeptide chains [44]. This component can be found in both Gram-negative and Gram-positive bacteria. In Gram-negative bacteria, the cell wall comprises 1 or 2 layers of peptidoglycan, while in Gram-positive bacteria, 10–40 layers are present [44,46].
There are different antimicrobials whose mechanisms of action inhibit cell wall synthesis: β-lactams, glycopeptides, and bacitracin, which is a polypeptide antibiotic [44,46,47]. Beta-lactams bind to transpeptidases (also called PBPs—penicillin-binding proteins), inhibiting the formation of peptide bonds between tetrapeptides that crosslink glycan chains, inactivating the PBPs, which results in the lysis of microorganisms [44,46]. Glycopeptides block cell wall synthesis by binding to the D-ala-D-ala terminus of the tetrapeptide chain, which also results in the inhibition of PBPs [44]. Bacitracin inactivates the membrane carrier, bactoprenol, responsible for the transport of peptidoglycan building blocks from the cytoplasm to the cell wall [47].

3.2. Inhibition of Protein Synthesis

Protein synthesis involves mRNA, tRNA, ribosomes, and other cytoplasmic factors and consists of three steps: initiation, elongation, and termination. The bacterial ribosome has two subunits, the 50S and 30S, each composed of rRNA and proteins [46].
There are several classes of antimicrobials that act to inhibit protein synthesis by binding to the 30S subunit (aminoglycosides, tetracyclines, and glycylcyclines) or the 50S subunit (macrolides, chloramphenicol, oxazolidinones, lincosamides, and streptogramin) [44,48,49,50]. Aminoglycosides act by binding with high affinity to the 16S rRNA of the 30S subunit. Thus, codons are misread when aminoacyl tRNA is delivered, resulting in erroneous protein synthesis. Consequently, the wrong amino acids are compiled into a polypeptide that is released, leading to apoptosis [44]. Tetracyclines, on the other hand, act through passive diffusion in the cell membrane by porin channels and reversibly bind to the 30S subunit, resulting in blocking the binding of the tRNA to the mRNA-ribosome complex [44]. Glycylcyclines are an antimicrobial class developed to overcome the mechanisms of resistance to tetracycline (ribosomal protection and efflux pumps). They bind to the 30S subunit with five times more affinity, inhibiting protein synthesis. On the other hand, glycylcyclines are not recognized by the tetracycline efflux transporter, exhibiting significant antibacterial activity [50].
Macrolides and oxazolidinones bind to the 23S rRNA of the 50S subunit and inhibit the process of translocation or transpeptidation of protein synthesis, inducing a premature separation of incomplete peptide chains. Chloramphenicol crosses the cell membrane and reversibly binds to the L16 protein of the 50S subunit, thus inhibiting the formation of peptide bonds and preventing the elongation of peptide chains [44].
Additionally, nitrofurans are bacteriostatic antimicrobials whose multiple mechanisms of action are not fully understood [51]. They inhibit the synthesis of proteins, DNA, and RNA [52]. Moreover, their wide mechanisms of action may explain the lack of acquired bacterial resistance to nitrofurans [51,52].
Very recently, streptothricin F has been revisited as a bactericidal antimicrobial effective against highly drug-resistant Gram-negative bacteria, namely carbapenem-resistant Enterobacterales (CRE), Acinetobacter baumannii, and Brucella abortus, as well as Mycobacterium tuberculosis. Streptothricin is a natural product mixture, currently referred to as nourseothricin. Its therapeutic use was abandoned due to its induced reversible kidney toxicity; however, new cytotoxic studies have shown that streptothricin F exhibits at least 10-fold lower toxicity than streptothricin D and nourseothricin, both in vitro and in vivo. Moreover, streptothricin F has an alternative and unique mechanism of action, interacting with the 30S subunit of the 70S ribosome [53].

3.3. Inhibition of Nucleic Acid Synthesis

Examples of antimicrobials that inhibit nucleic acid synthesis are ansamycins (e.g., rifamycin and rifampicin), fluoroquinolones, and nitroimidazoles (e.g., metronidazole) [44,46,54].
Ansamycins bind to the β-subunit of RNA polymerase, blocking RNA elongation and inhibiting RNA synthesis [44]. Fluoroquinolones act by inhibiting DNA gyrase and other topoisomerases, interfering with DNA replication [44,46]. Nitroimidazoles inhibit nucleic acid synthesis by forming nitroso radicals, which disrupt DNA. This class of antimicrobials is only effective against anaerobic bacteria, whose ferredoxin reduces them to active radicals [54].

3.4. Inhibition of Metabolic Pathways

Nitrogenous bases (purines and pyrimidines), formed through the folic acid pathway, are necessary for the synthesis of nucleic acids. This process is initiated with para-aminobenzoic acid (PABA), which is catalysed in dihydroflolic acid and subsequently in tetrahydrofolic acid, which is later used to synthesize nitrogenous bases [46].
Antimicrobials that inhibit folic acid synthesis are sulphonamides and trimethoprim [44]. Sulphonamides are structural analogues of PABA, competitively inhibiting the enzymatic conversion that leads to the production of dihydroflolic acid [44]. As for trimethoprim, it reversely inhibits the formation of tetrahydrofolic acid [44]. Used separately, trimethoprim and sulphonamides are bacteriostatic; however, combined, they seem to have a bactericidal effect [44].

3.5. Inhibition of Cell Membrane Function

Only a small class of antimicrobials act by inhibiting cell membrane function, the polymyxins. This class of lipopeptides consists of lipophilic detergent-type antimicrobials, which lyse cell membranes by destroying the lipopolysaccharide (LPS) layer [44].

4. Antimicrobial Resistance

Bacterial antimicrobial resistance may be natural or acquired. Natural resistance is either innate when constitutively expressed, or mediated if triggered by an antibiotic treatment. On the other hand, acquired resistance occurs through DNA mutation or via the transfer of genetic material between bacteria [44].
Bacteria can acquire antimicrobial resistance through genetic mutation, namely spontaneous mutation, hypermutation, and adaptive mutation. Spontaneous mutation can be driven by several factors, mainly errors in DNA replication, such as transitions, transversions, insertions and deletions, which are transmitted to the progeny. Hypermutation plays a crucial role in the evolution of antimicrobial resistance. Hypermutation is regulated by the SOS-inducible DNA polymerase IV. This mutation occurs in bacteria called hypermutators, as they have a greater affinity to undergo spontaneous mutations due to defects or repairs in DNA, or errors in the avoidance system. Therefore, hypermutators can quickly adapt to antimicrobials. Finally, adaptive mutation arises in non-diving bacteria, upon non-lethal selective pressure, such as nutrient conditions, or sub-inhibitory antimicrobial concentrations. This type of mutation is transient and can be reverted to the original condition in the absence of the pressure factor [45].
Antimicrobial resistance genes can be acquired by horizontal gene transfer between bacteria, either by conjugation, transformation, or transduction [21,55,56,57]. Conjugation (Figure 2) is the transient fusion between two bacteria, where the transfer of genetic material takes place from the donor to the recipient through conjugation pili. Transformation is the uptake of free genetic material, released by a donor bacterium, by a recipient bacterium. Finally, transduction is the transfer of resistant genes mediated by bacteriophages [21,55,57]. Among AMR gene transfer mechanisms, conjugation has been shown to play an important role in the transmission and dissemination of AMR in food [18].
The persistent use of antimicrobials, as well as misuse and self-medication, leads to the abovementioned acquired AMR. Moreover, the appearance of MDR, bacteria resistant to three or more antimicrobial classes, is a critical public health problem [2,22,33,58]. The treatment of infections caused by MDR bacteria poses a relevant clinical challenge since the increase in AMR leads to higher rates of therapeutic failures, relapses, longer hospitalizations, and worse clinical outcomes [3].
The increase in AMR triggers the need for surveillance of bacteria resistant to antimicrobials, which has been carried out in public health and food safety laboratories through Whole Genome Sequencing (WGS), enabling the description of the full AMR profile [21,59].
Bacterial resistance processes are divided into four biochemical mechanisms, which are highlighted in Figure 3 [44,45].

4.1. Antimicrobial Inactivation

Inactivation of the antimicrobial molecule occurs through the action of enzymes produced by resistant bacteria, such as β-lactamases and aminoglycoside-modifying enzymes [44,45,60]. Enzymes act on the antimicrobial molecule through hydrolysis, group transfer, or redox process. Hydrolysis is the process of destruction of the β-lactam ring of penicillin, cephalosporins, and carbapenems by β-lactamase-producing bacteria. Acyltransferases, phosphotransferases, and thioltransferases are examples of enzymes involved in hydrolysis, causing the destruction of the β-lactam ring and inhibiting the antimicrobial molecule binding to PBPs [44,45,60]. Group transfer, namely phosphoryl, acetyl, or adenyl group transfer to the antimicrobial active molecule, is considered the most effective mechanism of antimicrobial inactivation. An example of group transfer is acetylation on aminoglycosides, where enzymes alter hydroxyl or amino groups covalently, rendering antimicrobials inactive [44]. Finally, the redox process is the least studied mechanism, where antimicrobials are inactivated by oxidation or reduction [45].

4.2. Decreased Antimicrobial Penetration

Decreased antimicrobial penetration occurs through decreased cell wall permeability [44,45]. Gram-negative bacteria are intrinsically less permeable to certain antimicrobials than Gram-positive bacteria due to the large layer of LPS in the outer membrane of the cell wall that creates a permeability shield [44]. Hydrophilic molecules may penetrate the Gram-negative cell wall through porin proteins [45]. However, high-molecular-weight hydrophilic molecules, such as vancomycin, cannot pass through porins and are thus ineffective against Gram-negative bacteria [44].
Some bacteria are able to downregulate the expression of porins or even replace them with non-selective channels, decreasing the cell wall permeability and becoming thus resistant to some antimicrobials [44]. Hydrophilic molecules, such as β-lactams, tetracyclines, and some fluoroquinolones, are greatly affected by changes in the permeability of the outer membrane [44].

4.3. Activation of the Efflux Pump

The efflux system consists of energy-dependent membrane transport systems that pump a wide range of molecules [60]. In this transport system, there are efflux pumps, which are transport proteins that are located mostly in the bacterial cytoplasmic membrane [45,60]. These proteins transport nutrients and excrete cellular toxic compounds through the proton matrix force [45].
Efflux pumps can be specific to a particular antimicrobial or multi-resistant efflux pumps capable of excreting various antimicrobials [44,45,60]. The main families of efflux pumps are ATP-binding cassettes (ABC), small multidrug resistance (SMR), multidrug and toxic component extrusion (MATE), resistance-nodulation cell division (RND), and large facilitator superfamily (MFS) [44,60]. This mechanism confers resistance to macrolides, β-lactams, fluoroquinolones, 4th generation cephalosporins, carbapenems, tetracyclines, and oxazolidines [44,45].

4.4. Target Modification

The modification of the antimicrobial target is one of the most common resistance mechanisms. For β-lactams, changes may occur either in the composition or the amount of PBPs. Thus, the amount of antimicrobial that can bind to the target is affected by the change in the number of PBPs, while a structural modification decreases or completely prevents the binding of the molecules [44,60]. Another method is the production of alternative proteins that adopt the role of the bacterium’s native protein, resulting in antimicrobial resistance [45]. Moreover, modification of ribosomes or the peptidoglycan precursor can also occur. Ribosome modification consists of ribosome methylation, commonly mediated by erm gene products, which can be constitutive or inducible. This modification results in resistance to macrolides, lincosamides, and streptogramin B [60]. Regarding the modification of the peptidoglycan precursor, in the case of resistance to glycopeptides, it occurs through an amino acid substitution. The change occurs at the end of the D-alanyl-D-alanine dipeptide that is found at the terminals of the tetrapeptide [60].

5. Antimicrobial Resistance and Farm-to-Fork Transmission

Antimicrobial resistance in foods is considered a food safety issue but also a relevant public health problem. Furthermore, awareness of the prevalence of foodborne pathogenic bacterial strains resistant to antimicrobials is of the utmost importance [21,33].
The presence of bacteria resistant to antimicrobials in foods of animal origin has increased dramatically in recent years [33,61]. Moreover, bacteria have the ability to evolve and gain resistance to new antimicrobials [22,33,61]. Therefore, humans are highly exposed to AMR bacteria through food consumption [21,22,61,62], mainly due to the use of antimicrobials in the livestock sector [21,33,61,62]. Thus, the food chain has a high impact on the transmission of AMR, as food is not sterile and usually gets microbiological contamination via cross-contamination or recontamination throughout manufacturing. So, the food chain is considered a driver for the transmission of AMR bacteria [22,61,63,64].
The transfer of AMR bacteria from food products to humans occurs by consumption, followed by the horizontal transfer of resistance genes in the human gut [21,22,65]. Recently, several studies have studied the microbiome along the production chain to assess the AMR genes present in food samples [61,66]. These microbiome studies may contribute to the production of safer meat and meat products within the framework of One Health [66].
Therefore, several policy objectives have been considered to reduce antimicrobials in foods, such as a 50% reduction in the sales of antimicrobials for farmed animals and in aquaculture until 2030, aligned with the Farm-to-Fork Strategy of the European Green Deal [67]. Nevertheless, this may not be enough to effectively control AMR [68].

5.1. The Role of Meat in the Transmission of Antimicrobial Resistance

Meat and meat products are an important source of protein, vitamins, and minerals in the human diet and, in some countries, play an important role in gastronomic culture [69,70,71,72]. From the nutritional point of view, meat is considered a valuable source of protein, due to its amino acid composition, along with the presence of iron, zinc, and vitamins B12 and D, as well as other micronutrients [69,71]. The consumption of meat has been increasingly growing and is expected to reach between 460 and 570 million tons per year by 2050 [73,74].
Along with the increase in meat consumption, the demand for meat products has also grown, mainly due to their sensory properties and the opportunity to use parts of the carcass that cannot be used for fresh consumption [69,72]. Another advantage of meat products is their extended shelf-life. Meanwhile, meat products are also a vehicle for microorganisms, with either a beneficial, neutral, or harmful effect on health [72].
Within the animal industry, the rearing and consumption of pork meat have grown enormously in recent years, mainly because pork is a high-quality, low-cost animal protein [70]. However, pigs are considered one of the biggest reservoirs of AMR [65,75,76,77], mainly due to the inappropriate use of antimicrobials. In some countries, the administration of antimicrobials to promote animal growth is still allowed [78,79,80]. Excessive use of antimicrobials applies a selective pressure that leads to the development of antimicrobial-resistant bacterial populations, which may later be transmitted to humans [79].
Every step of the pig value chain, whether feeding, slaughtering, or processing, has the potential to affect human and animal health [38]. Moreover, there are two main sources of contamination in a pig slaughterhouse: the microorganisms carried on the pig’s skin and those from the evisceration step [81]. Mitigation measures along the food production chain may include enhanced disinfection procedures in the above-mentioned contamination-source areas in order to reduce the risk to food safety and consumer health due to the spread of antibiotic and virulence determinants to end products and the environment [81].
Hypervirulent clonal complexes (CCs) of Listeria monocytogenes were found in pig tonsils, showing the potential risk of pigs as source of isolates causing human listeriosis [82]. Moreover, a broad distribution of CC was observed along the whole pig production chain, suggesting multiple sources of entry [82].
Food contamination is the main cause of foodborne illnesses in both developed and developing countries [31]. Moreover, farm-to-fork AMR transmission is an additional food safety concern [83]. Considering the estimation that AMR will cause about 10 million deaths per year and cost US$100 trillion by 2050 [31,84,85], it is mostly relevant to control food contamination throughout the whole value chain.
Due to the impact of AMR and the fact that meat and meat products are highly consumed, there has been an increase in studies to evaluate the quality and safety of this type of food [72,86,87,88], including the search for antimicrobial-resistant pathogens in food (for example, Staphylococcus aureus, Escherichia coli, and Salmonella spp.), the associated antimicrobial resistance genes (Table 1), and the possibility of transmission to humans through consumption [86,89,90,91,92,93,94,95].
The microbiota of meat and meat products includes not only foodborne pathogens but also spoilage and technological microorganisms, which may all be responsible for farm-to-fork transmission of AMR [4,83]. For example, technological microbiota like coagulase-negative staphylococci can harbour antimicrobial resistance genes by acquiring them from other bacterial genera, normally pathogenic bacteria through horizontal gene transfer [105,106].
Additionally, there are studies that confirm the hypothesis of foodborne bacteria transferring antimicrobial resistance genes to the human gut microbiota [89,90,106,107,108,109]. Cao et al. (2022) worked with samples from 21 volunteers and pig and poultry carcasses and detected the presence of antimicrobial resistance genes (AMRGs) both in humans and food animals, conferring resistance to several antimicrobial classes: vancomycin, tetracycline and macrolides [90]. Moreover, this study showed that approximately 40% of AMRGs were shared between humans and pork, and 24.7% were shared between humans and poultry [90]. Bouchami et al. (2020) studied pigs, slaughter workers, and food contact surfaces. Staphylococcus aureus was selectively isolated from 41% of samples, 55% of which harboured the SCCmec type V cassette (methicillin-resistant S. aureus—MRSA), conferring resistance mainly to β-lactams, tetracycline, clindamycin, erythromycin, gentamicin and chloramphenicol [89]. These authors compared isolates from different sources and suggested the dissemination of MRSA from the pig production chain to humans. Lawal et al. (2021) evaluated Staphylococcus saprophyticus from human and slaughterhouse samples (equipment, pork meat, workers’ hands, and pigs’ rectum) [106]. The authors found AMRGs conferring resistance mainly to biocides (qaac) and trimethoprim (dfrG), both in foodborne and human isolates. L. monocytogenes was isolated from 12.5% of ready-to-eat meat-based products (RTEMBP), and 20% of the samples were considered MDR (resistant to gentamicin, meropenem, benzylpenicillin, quinupristin-dalfopristin, rifampin, sulphamethoxazole-trimethoprim, and tetracycline) [107]. The authors also detected a high similarity between RTEMBP and human clinical isolates.
These facts highlight the suitability of the One Health approach to control the AMR transmission process between food animals and humans.

5.2. Antimicrobial Resistance in Staphylococcus aureus

Staphylococcus aureus is an opportunistic, Gram-positive, round-shaped, facultative anaerobic pathogen that can often be found in the natural microbiota of both the nose and skin [25,110,111,112,113]. S. aureus is responsible for several life-threatening infections, such as endocarditis, toxic shock syndrome, and osteomyelitis [25,114,115]. S. aureus can also be found in foods (raw or ready-to-eat foods) due to contamination through the handling process, food-producing animals, and food contact surfaces [25,111,113]. High S. aureus load in food may cause food poisoning [25,111,113,115]. Moreover, S. aureus has been considered one of the most relevant microbiological hazards in meat and meat products because of their strong evidence association with foodborne outbreaks [116].
The excessive use of antimicrobials in the livestock sector led to the emergence of multi-resistant S. aureus in the food [30]. The most studied S. aureus is methicillin-resistant S. aureus (MRSA), because of the few effective treatments against infections with these strains [114].
The resistance to antimicrobials by S. aureus is due to mechanisms of intrinsic resistance, resistance mutations, or the acquisition of resistance mechanisms. Therefore, it is necessary to characterize the acquired resistance mechanisms through whole genome sequencing [111,117]. Methicillin-resistant S. aureus is considered multidrug-resistant bacteria, as resistance to almost all β-lactams, vancomycin, and fluoroquinolones has been reported [114,118]. MRSA resistance is due to horizontal transfer of genes and mobile genetic elements, such as the mobile staphylococcal cassette chromosome (SCCmec) that harbours the mecA, mecB, and mecC genes [25,111,119]. Regarding vancomycin resistance, it is conferred by the horizontal transfer of the vanA gene from Enterococcus spp. to S. aureus [120].
Evidence of foodborne transmission of MRSA has been reported by several authors [121,122]. Similarly, Bonardi et al. (2022) found a genetic relationship between swine and human isolates, although no direct epidemiological link was demonstrated [123].

5.3. Antimicrobial Resistance in ESBL-Producing Enterobacteriaceae

Enterobacteriaceae is a family of Gram-negative bacilli, facultative anaerobes, responsible for various community-acquired and nosocomial infections, such as urinary, lower respiratory tract and bloodstream infections [124,125,126]. The natural habitat of enterobacteria is the gut of humans and animals [28]. In recent years, Escherichia coli and Klebsiella pneumoniae, both ESBL-producing enterobacteria, have been the main species associated with nosocomial infections [28,124,127,128]. ESBL-producing enterobacteria are a group of bacteria consisting of K. pneumoniae, E. coli, Enterobacter spp, Proteus spp., Serratia spp., Providencia spp., Salmonella spp., and Morganella morganii, which are resistant to a wide range of β-lactams [28]. Due to their MDR resistance profile, ESBL enterobacteria are considered a critical priority in the WHO “List of Priority Pathogens” [129], which represents a public health problem, also being detected in the livestock and food sectors [28,130].
The extensive resistance of enterobacteria to β-lactams is due to the widespread use of these antimicrobials. ESBL are bacterial enzymes that confer resistance to broad-spectrum penicillin, among other β-lactams, like third-generation cephalosporines [28,125,131]. A specific ESBL produced by enterobacteria is AmpC β-lactamase, which is capable of hydrolysing penicillins, 1st to 3rd generation cephalosporins, cephamycins, and beta-lactamase inhibitors [132,133,134,135]. Additionally, metallo-beta-lactamases confer resistance to carbapenems [134,135].
Furthermore, enterobacteria often harbour mcr genes that confer resistance to colistin [130,132,136]. Recently, colistin-resistant enterobacteria have been reported on a large scale [124,137]. This resistance is due to a lower binding affinity for colistin, through the modification of the lipid A component of LPS. mcr genes are found in plasmids, accelerating the transfer of resistance between bacterial strains [124].
Identical strains of ESBL-E. coli were isolated from both healthy humans and swine [138]. Moreover, common transposable elements were found in ESBL-E. coli isolates from human and non-human sources [139].

5.4. Antimicrobial Resistance in Vancomycin-Resistant Enterococcus spp.

Lactic acid bacteria (LAB) are Gram-positive cocci or bacilli, non-spore-forming, anaerobic, catalase-negative, and able to ferment glucose, resulting in the production of lactic acid, CO2, and ethanol [140,141]. Although most LABs are beneficial, some species are opportunistic pathogens for animals and humans, as is the case of some enterococci commonly found in the gastrointestinal tract [29,140,142]. Within LAB, E. faecium and E. faecalis are the most problematic species, responsible for nosocomial infections such as bloodstream, urinary tract, endocardium, and skin infections [26,141,143,144,145]. In recent years, infections with antimicrobial-resistant enterococci have been reported due to their intrinsic resistance to vancomycin and penicillin and both intrinsic and acquired resistance to aminoglycosides and macrolides [26,146,147,148]. Furthermore, a small percentage of linezolid-resistant enterococci, as well as enterococci with a low susceptibility to both linezolid and tedizolid, have been reported [26,147,148,149].
However, vancomycin-resistant enterococci are the main opportunistic pathogens, being classified as high-priority pathogens by the WHO [129]. They may be found in foods due to their ability to adapt to various environmental conditions, such as the production and storage environments for ready-to-eat foods [29,150]. Bearing in mind that enterococci have very plastic genomes capable of acquiring and transferring resistance to antimicrobials, enterococcal infections are very difficult to treat because enterococci easily become multidrug-resistant [142]. Enterococci resistance to vancomycin is due to the acquisition of van genes. While vanA is widely used in the identification of mobile genetic elements, vanC1, vanC2, and vanC3 genes are responsible for the intrinsic resistance of enterococci [147,150]. When vancomycin-resistant enterococci also exhibit resistance to ampicillin, treatment of infections is usually limited to the use of last-resort antimicrobials such as linezolid, tigecycline, and daptomycin [146,151]. However, enterococci resistant to oxazolidinones (linezolid, tedizolid) have arisen, which results from the acquisition of transferable plasmid genes, namely cfr, cfr (B), cfr (C), and optrA, mutations in the 23S rRNA genes, and mutations in the ribosomal proteins L3 and L4 genes [146,149].
Identical strains of Enterococcus faecalis resistant to gentamicin have been found in patients and pigs in Denmark [152].

5.5. Antimicrobial Resistance in ESKAPE Bacteria

ESKAPE bacteria are considered one of the greatest dangers in modern medicine, because they are MDR bacteria, often causing nosocomial infections [27], which are one of the main causes of morbidity and mortality across the world [2,153]. All bacteria that belong to this group are opportunistic pathogens, showing several antimicrobial resistance mechanisms, such as target modification, enzymatic inactivation, and mechanical protection (biofilm formation) [27,154]. ESKAPE bacteria belong to the list of WHO pathogens, Priority 1 (Critical antibiotic resistance), and Priority 2 (High antibiotic resistance) levels [129]. Gram-negative ESKAPE bacteria belong to Priority 1 (Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter spp.), while Gram-positive ESKAPE bacteria are Priority 2 (Enterococcus faecium and Staphylococcus aureus) [153]. Among the resistance mechanisms of ESKAPE bacteria, biofilm formation has been the focus of greatest concern, with biofilm acting as a physical barrier to host immune mechanisms and antimicrobial molecules. In fact, biofilms can even protect antimicrobial-tolerant bacteria [2].
Enterococcus faecium and Staphylococcus aureus are two clinically relevant Gram-positive bacteria, frequently responsible for nosocomial infections [27].
Klebsiella pneumoniae is a Gram-negative bacterium of the Enterobacteriaceae family. These are encapsulated rod-shaped, facultative anaerobes found in the gastrointestinal tract, responsible for several infections, such as urinary infections and pneumonia. The resistance of K. pneumoniae to antimicrobials is due to the production of extended-spectrum β-lactamases (ESBL), which putatively confer resistance to β-lactams, cephalosporines, monobactams and carbapenems [27,153,155].
Acinetobacter baumannii is a Gram-negative coccobacillus, which is strictly aerobic and non-fermentative. It is frequently found in hospital environments, causing bloodstream infections, among others. This bacterium has a high ability to survive on surfaces due to biofilm production, produces ESBL, its genome evolves rapidly; and it can acquire AMR genes under selective pressure [27,153,155].
Pseudomonas aeruginosa is a Gram-negative rod-shaped, strictly aerobic, encapsulated bacterium considered an opportunistic pathogen. It can cause sepsis, pneumonia, and other difficult-to-treat infections. Its resistance to antimicrobials is due to the acquisition of mobile resistance genes, biofilm formation, and expression of porins and efflux pumps, resulting in resistance to colistin, chloramphenicol, tetracycline, β-lactams, rifampin, and trimethoprim-sulfamethoxazole [27,153,155].
Enterobacter is a Gram-negative bacillus of the Enterobacteriaceae family, a facultative anaerobe, belonging to the human microbiota. It is an opportunistic pathogen that causes infections, such as pneumonia, sepsis, and urinary tract infections, among others. Enterobacter is intrinsically resistant to ampicillin, amoxicillin, first-generation cephalosporins, and cefoxitin due to the presence of a constitutive AmpC β-lactamase [156]. Since they produce ESBL and carbapenemases, they further harbour various resistance bla genes (blaNDM, blaOXA, blaKPC, blaVIM, blaCTX-M, blaIMP, and blaTEM) [27,153,155].

6. Conclusions

Despite the guidelines that have been implemented worldwide, and especially in Europe, within the scope of antimicrobial stewardship, and the efforts made by professionals involved in human and animal health nowadays, AMR is still a recurrent global problem responsible for high morbidity rates, leading to thousands of deaths each year.
Besides being a problem for causing foodborne infections or intoxications, foodborne bacteria can also carry antimicrobial resistance genes. Specifically, in pork meat products, antimicrobial resistance genes have been detected for ampicillin, chloramphenicol, clindamycin, gentamycin, kanamycin, nitrofurantoin, quinolone, streptomycin, tetracycline, trimethoprim, and tylosin. Moreover, in the human digestive tract, transfer of resistance genes to indigenous gut bacteria may occur. Additionally, identical isolates and highly similar antimicrobial resistance genes were detected in meat and meat products, other ready-to-eat meat-based food, and human clinical isolates.
Therefore, AMR foodborne (from foods to humans) transmission, or “farm-to-fork” transmission, has been reported and should be of the utmost concern, particularly in the case of pork meat and meat products. Furthermore, from a One Health perspective, different disciplines are necessary and should be integrated to control the problem of AMR globally.

Author Contributions

Conceptualization, M.L.; writing—original draft preparation, S.C.; writing—review and editing, M.C.Q. and M.L.; supervision, M.L.; funding acquisition, M.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by National Funds through FCT—Foundation for Science and Technology under the Project UIDB/05183/2020. S. Conceição acknowledges a PhD fellowship from FCT under the reference UI/BD/153510/2022.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Scott, H.M.; Acuff, G.; Bergeron, G.; Bourassa, M.W.; Gill, J.; Graham, D.W.; Kahn, L.H.; Morley, P.S.; Salois, M.J.; Simjee, S.; et al. Critically important antibiotics: Criteria and approaches for measuring and reducing their use in food animal agriculture. Ann. N. Y. Acad. Sci. 2019, 1441, 8–16. [Google Scholar] [CrossRef] [PubMed]
  2. Mulani, M.S.; Kamble, E.E.; Kumkar, S.N.; Tawre, M.S.; Pardesi, K.R. Emerging strategies to combat ESKAPE pathogens in the era of antimicrobial resistance: A review. Front. Microbiol. 2019, 10, 539. [Google Scholar] [CrossRef] [PubMed]
  3. Gargiullo, L.; Del Chierico, F.; D’Argenio, P.; Putignani, L. Gut microbiota modulation for multidrug-resistant organism decolonization: Present and future perspectives. Front. Microbiol. 2019, 10, 1704. [Google Scholar] [CrossRef] [PubMed]
  4. Tong, J.; Zhang, Z.; Wu, Q.; Huang, Z.; Malakar, P.K.; Chen, L.; Liu, H.; Pan, Y.; Zhao, Y. Antibacterial peptides from seafood: A promising weapon to combat bacterial hazards in food. Food Control 2021, 125, 108004. [Google Scholar] [CrossRef]
  5. Queiroga, C.; Andrade, N.; Laranjo, M. Antimicrobial action of propolis extracts against staphylococci. In Understanding Microbial Pathogens Current Knowledge and Educational Ideas on Antimicrobial Research; Formatex Research Center: Badajoz, Spain, 2018; pp. 1–8. [Google Scholar]
  6. Queiroga, M.C.; Laranjo, M.; Andrade, N.; Marques, M.; Costa, A.R.; Antunes, C.M. Antimicrobial, Antibiofilm and Toxicological Assessment of Propolis. Antibiotics 2023, 12, 347. [Google Scholar] [CrossRef]
  7. Queiroga, M.C.; Pinto Coelho, M.; Arantes, S.M.; Potes, M.E.; Martins, M.R. Antimicrobial Activity of Essential Oils of Lamiaceae Aromatic Spices Towards Sheep mastitis-Causing Staphylococcus aureus and Staphylococcus epidermidis. J. Essent. Oil Bear. Plants 2018, 21, 1155–1165. [Google Scholar] [CrossRef]
  8. Laranjo, M.; Fernández-León, A.M.; Agulheiro-Santos, A.C.; Potes, M.E.; Elias, M. Essential oils of aromatic and medicinal plants play a role in food safety. J. Food Process. Preserv. 2019, 46, e14278. [Google Scholar] [CrossRef]
  9. Mohammadi Gheisar, M.; Kim, I.H. Phytobiotics in poultry and swine nutrition—A review. Ital. J. Anim. Sci. 2017, 17, 92–99. [Google Scholar] [CrossRef]
  10. Dellit, T.H.; Owens, R.C.; McGowan, J.E.; Gerding, D.N.; Weinstein, R.A.; Burke, J.P.; Huskins, W.C.; Paterson, D.L.; Fishman, N.O.; Carpenter, C.F.; et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America Guidelines for Developing an Institutional Program to Enhance Antimicrobial Stewardship. Clin. Infect. Dis. 2007, 44, 159–177. [Google Scholar] [CrossRef]
  11. Sudatip, D.; Chasiri, K.; Kritiyakan, A.; Phanprasit, W.; Thinphovong, C.; Tiengrim, S.; Thamlikitkul, V.; Abdallah, R.; Baron, S.A.; Rolain, J.M.; et al. A One Health approach to assessing occupational exposure to antimicrobial resistance in Thailand: The FarmResist project. PLoS ONE 2021, 16, e0245250. [Google Scholar] [CrossRef]
  12. Nations, U. Transforming Our World: The 2030 Agenda for Sustainable Development. 2015. Available online: https://sdgs.un.org/publications/transforming-our-world-2030-agenda-sustainable-development-17981 (accessed on 13 July 2023).
  13. Huan, Y.; Yu, Y.; Liang, T.; Burgman, M. A method for assessing the impacts of an international agreement on regional progress towards Sustainable Development Goals. Sci. Total Environ. 2021, 785, 147336. [Google Scholar] [CrossRef]
  14. Khaled, R.; Ali, H.; Mohamed, E.K.A. The Sustainable Development Goals and corporate sustainability performance: Mapping, extent and determinants. J. Clean. Prod. 2021, 311, 127599. [Google Scholar] [CrossRef]
  15. Pohlmann, C.R.; Scavarda, A.J.; Alves, M.B.; Korzenowski, A.L. The role of the focal company in sustainable development goals: A Brazilian food poultry supply chain case study. J. Clean. Prod. 2020, 245, 118798. [Google Scholar] [CrossRef]
  16. Hou, J.; Long, X.; Wang, X.; Li, L.; Mao, D.; Luo, Y.; Ren, H. Global trend of antimicrobial resistance in common bacterial pathogens in response to antibiotic consumption. J. Hazard. Mater. 2023, 442, 130042. [Google Scholar] [CrossRef]
  17. Michael, A.; Kelman, T.; Pitesky, M. Overview of quantitative methodologies to understand antimicrobial resistance via minimum inhibitory concentration. Animals 2020, 10, 1405. [Google Scholar] [CrossRef] [PubMed]
  18. Mouiche, M.M.M.; Moffo, F.; Akoachere, J.F.T.K.; Okah-Nnane, N.H.; Mapiefou, N.P.; Ndze, V.N.; Wade, A.; Djuikwo-Teukeng, F.F.; Toghoua, D.G.T.; Zambou, H.R.; et al. Antimicrobial resistance from a one health perspective in Cameroon: A systematic review and meta-analysis. BMC Public Health 2019, 19, 1135. [Google Scholar] [CrossRef]
  19. Bordier, M.; Binot, A.; Pauchard, Q.; Nguyen, D.T.; Trung, T.N.; Fortané, N.; Goutard, F.L. Antibiotic resistance in Vietnam: Moving towards a One Health surveillance system. BMC Public Health 2018, 18, 1136. [Google Scholar] [CrossRef]
  20. Sun, Q.; Wang, Y.; Hulth, A.; Xiao, Y.; Nilsson, L.E.; Li, X.; Bi, Z.; Liu, Y.; Yin, H.; Luo, Y.; et al. Study protocol for One Health data collections, analyses and intervention of the Sino-Swedish integrated multisectoral partnership for antibiotic resistance containment (IMPACT). BMJ Open 2018, 8, e017832. [Google Scholar] [CrossRef]
  21. Collineau, L.; Boerlin, P.; Carson, C.A.; Chapman, B.; Fazil, A.; Hetman, B.; McEwen, S.A.; Jane Parmley, E.; Reid-Smith, R.J.; Taboada, E.N.; et al. Integrating whole-genome sequencing data into quantitative risk assessment of foodborne antimicrobial resistance: A review of opportunities and challenges. Front. Microbiol. 2019, 10, 1107. [Google Scholar] [CrossRef]
  22. Pérez-Rodríguez, F.; Mercanoglu Taban, B. A State-of-Art Review on Multi-Drug Resistant Pathogens in Foods of Animal Origin: Risk Factors and Mitigation Strategies. Front. Microbiol. 2019, 10, 2091. [Google Scholar] [CrossRef]
  23. Zhuo, A.; Labbate, M.; Norris, J.M.; Gilbert, G.L.; Ward, M.P.; Bajorek, B.V.; Degeling, C.; Rowbotham, S.J.; Dawson, A.; Nguyen, K.A.; et al. Opportunities and challenges to improving antibiotic prescribing practices through a One Health approach: Results of a comparative survey of doctors, dentists and veterinarians in Australia. BMJ Open 2018, 8, e020439. [Google Scholar] [CrossRef] [PubMed]
  24. Jiang, X.; Cui, X.; Xu, H.; Liu, W.; Tao, F.; Shao, T.; Pan, X.; Zheng, B. Whole genome sequencing of extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli isolated from a wastewater treatment plant in China. Front. Microbiol. 2019, 10, 1797. [Google Scholar] [CrossRef] [PubMed]
  25. Li, H.; Andersen, P.S.; Stegger, M.; Sieber, R.N.; Ingmer, H.; Staubrand, N.; Dalsgaard, A.; Leisner, J.J. Antimicrobial Resistance and Virulence Gene Profiles of Methicillin-Resistant and -Susceptible Staphylococcus aureus From Food Products in Denmark. Front. Microbiol. 2019, 10, 2681. [Google Scholar] [CrossRef]
  26. Wen, Z.; Shang, Y.; Xu, G.; Pu, Z.; Lin, Z.; Bai, B.; Chen, Z.; Zheng, J.; Deng, Q.; Yu, Z. Mechanism of Eravacycline Resistance in Clinical Enterococcus faecalis Isolates from China. Front. Microbiol. 2020, 11, 916. [Google Scholar] [CrossRef] [PubMed]
  27. Cieślik, M.; Bagińska, N.; Górski, A.; Jończyk-Matysiak, E. Animal models in the evaluation of the effectiveness of phage therapy for infections caused by gram-negative bacteria from the ESKAPE group and the reliability of its use in humans. Microorganisms 2021, 9, 206. [Google Scholar] [CrossRef] [PubMed]
  28. Collis, R.M.; Burgess, S.A.; Biggs, P.J.; Midwinter, A.C.; French, N.P.; Toombs-Ruane, L.; Cookson, A.L. Extended-spectrum beta-lactamase-producing enterobacteriaceae in dairy farm environments: A New Zealand perspective. Foodborne Pathog. Dis. 2019, 16, 5–22. [Google Scholar] [CrossRef]
  29. Igbinosa, E.O.; Beshiru, A. Antimicrobial resistance, virulence determinants, and biofilm formation of Enterococcus species from ready-to-eat seafood. Front. Microbiol. 2019, 10, 728. [Google Scholar] [CrossRef]
  30. Lopes, E.; Conceição, T.; Poirel, L.; de Lencastre, H.; Aires-De-Sousa, M. Epidemiology and antimicrobial resistance of methicillin-resistant Staphylococcus aureus isolates colonizing pigs with different exposure to antibiotics. PLoS ONE 2019, 14, e0225497. [Google Scholar] [CrossRef]
  31. Aaliya, B.; Valiyapeediyekkal Sunooj, K.; Navaf, M.; Parambil Akhila, P.; Sudheesh, C.; Ahmed Mir, S.; Sabu, S.; Sasidharan, A.; Theingi Hlaing, M.; George, J. Recent trends in bacterial decontamination of food products by hurdle technology: A synergistic approach using thermal and non-thermal processing techniques. Food Res. Int. 2021, 147, 110514. [Google Scholar] [CrossRef]
  32. Cooper, A.L.; Low, A.J.; Koziol, A.G.; Thomas, M.C.; Leclair, D.; Tamber, S.; Wong, A.; Blais, B.W.; Carrillo, C.D. Systematic Evaluation of Whole Genome Sequence-Based Predictions of Salmonella Serotype and Antimicrobial Resistance. Front. Microbiol. 2020, 11, 549. [Google Scholar] [CrossRef]
  33. Thapa, S.P.; Shrestha, S.; Anal, A.K. Addressing the antibiotic resistance and improving the food safety in food supply chain (farm-to-fork) in Southeast Asia. Food Control 2020, 108, 106809. [Google Scholar] [CrossRef]
  34. Adisasmito, W.B.; Almuhairi, S.; Behravesh, C.B.; Bilivogui, P.; Bukachi, S.A.; Casas, N.; Becerra, N.C.; Charron, D.F.; Chaudhary, A.; Ciacci Zanella, J.R.; et al. One Health: A new definition for a sustainable and healthy future. PLoS Pathog. 2022, 18, e1010537. [Google Scholar] [CrossRef]
  35. What Is One Health? Available online: https://www.onehealthcommission.org/en/why_one_health/what_is_one_health/ (accessed on 6 June 2023).
  36. Mitchell, M.E.V.; Alders, R.; Unger, F.; Nguyen-Viet, H.; Le, T.T.H.; Toribio, J.A. The challenges of investigating antimicrobial resistance in Vietnam—What benefits does a One Health approach offer the animal and human health sectors? BMC Public Health 2020, 20, 213. [Google Scholar] [CrossRef] [PubMed]
  37. Baquero, F.; Coque, T.M.; Martínez, J.L.; Aracil-Gisbert, S.; Lanza, V.F. Gene Transmission in the One Health Microbiosphere and the Channels of Antimicrobial Resistance. Front. Microbiol. 2019, 10, 2892. [Google Scholar] [CrossRef] [PubMed]
  38. Qian, J.; Wu, Z.; Zhu, Y.; Liu, C. One Health: A holistic approach for food safety in livestock. Sci. One Health 2022, 1, 100015. [Google Scholar] [CrossRef]
  39. Netea, M.G.; Dominguez-Andres, J.; Barreiro, L.B.; Chavakis, T.; Divangahi, M.; Fuchs, E.; Joosten, L.A.B.; van der Meer, J.W.M.; Mhlanga, M.M.; Mulder, W.J.M.; et al. Defining trained immunity and its role in health and disease. Nat. Rev. Immunol. 2020, 20, 375–388. [Google Scholar] [CrossRef]
  40. Byrne, K.A.; Tuggle, C.K.; Loving, C.L. Differential induction of innate memory in porcine monocytes by beta-glucan or bacillus Calmette-Guerin. Innate Immun. 2021, 27, 448–460. [Google Scholar] [CrossRef]
  41. Campos, M.; Capilla, R.; Naya, F.; Futami, R.; Coque, T.; Moya, A.; Fernandez-Lanza, V.; Cantón, R.; Sempere, J.M.; Llorens, C.; et al. Simulating Multilevel Dynamics of Antimicrobial Resistance in a Membrane Computing Model. mBio 2019, 10, e02460-18. [Google Scholar] [CrossRef]
  42. Bizzaro, G.; Vatland, A.K.; Pampanin, D.M. The One-Health approach in seaweed food production. Environ. Int. 2022, 158, 106948. [Google Scholar] [CrossRef]
  43. Jeleff, M.; Lehner, L.; Giles-Vernick, T.; Dückers, M.L.A.; Napier, D.; Jirovsky-Platter, E.; Kutalek, R. Vulnerability and One Health assessment approaches for infectious threats from a social science perspective: A systematic scoping review. Lancet Planet. Health 2022, 6, e682–e693. [Google Scholar] [CrossRef]
  44. Uddin, T.M.; Chakraborty, A.J.; Khusro, A.; Zidan, B.M.R.M.; Mitra, S.; Emran, T.B.; Dhama, K.; Ripon, M.K.H.; Gajdács, M.; Sahibzada, M.U.K.; et al. Antibiotic resistance in microbes: History, mechanisms, therapeutic strategies and future prospects. J. Infect. Public Health 2021, 14, 1750–1766. [Google Scholar] [CrossRef] [PubMed]
  45. Pulingam, T.; Parumasivam, T.; Gazzali, A.M.; Sulaiman, A.M.; Chee, J.Y.; Lakshmanan, M.; Chin, C.F.; Sudesh, K. Antimicrobial resistance: Prevalence, economic burden, mechanisms of resistance and strategies to overcome. Eur. J. Pharm. Sci. 2022, 170, 106103. [Google Scholar] [CrossRef] [PubMed]
  46. Upmanyu, N.; Malviya, V.N. Antibiotics: Mechanisms of action and modern challenges. In Microorganisms for Sustainable Environment and Health; Elsevier: Amsterdam, The Netherlands, 2020; pp. 367–382. [Google Scholar]
  47. Singh, S.P.; Qureshi, A.; Hassan, W. Mechanisms of action by antimicrobial agents: A review. McGill J. Med. 2021, 19. [Google Scholar] [CrossRef]
  48. Lin, J.; Zhou, D.; Steitz, T.A.; Polikanov, Y.S.; Gagnon, M.G. Ribosome-Targeting Antibiotics: Modes of Action, Mechanisms of Resistance, and Implications for Drug Design. Annu. Rev. Biochem. 2018, 87, 451–478. [Google Scholar] [CrossRef]
  49. Johnston, N.; Mukhtar, T.; Wright, G. Streptogramin Antibiotics: Mode of Action and Resistance. Curr. Drug Targets 2002, 3, 335–344. [Google Scholar] [CrossRef]
  50. Zuckerman, J.M.; Qamar, F.; Bono, B.R. Review of Macrolides (Azithromycin, Clarithromycin), Ketolids (Telithromycin) and Glycylcyclines (Tigecycline). Med. Clin. N. Am. 2011, 95, 761–791. [Google Scholar] [CrossRef]
  51. Wijma, R.A.; Huttner, A.; Koch, B.C.P.; Mouton, J.W.; Muller, A.E. Review of the pharmacokinetic properties of nitrofurantoin and nitroxoline. J. Antimicrob. Chemother. 2018, 73, 2916–2926. [Google Scholar] [CrossRef]
  52. Vallée, M.; Harding, C.; Hall, J.; Aldridge, P.D.; Tan, A. Exploring the in situ evolution of nitrofurantoin resistance in clinically derived uropathogenic Escherichia coli isolates. J. Antimicrob. Chemother. 2022, 78, 373–379. [Google Scholar] [CrossRef]
  53. Morgan, C.E.; Kang, Y.-S.; Green, A.B.; Smith, K.P.; Dowgiallo, M.G.; Miller, B.C.; Chiaraviglio, L.; Truelson, K.A.; Zulauf, K.E.; Rodriguez, S.; et al. Streptothricin F is a bactericidal antibiotic effective against highly drug-resistant gram-negative bacteria that interacts with the 30S subunit of the 70S ribosome. PLoS Biol. 2023, 21, e3002091. [Google Scholar] [CrossRef]
  54. Edwards, D.I. Nitroimidazole drugs-action and resistance mechanisms I. Mechanisms of action. J. Antimicrob. Chemother. 1993, 31, 9–20. [Google Scholar] [CrossRef]
  55. De Lucia, A.; Card, R.M.; Duggett, N.; Smith, R.P.; Davies, R.; Cawthraw, S.A.; Anjum, M.F.; Rambaldi, M.; Ostanello, F.; Martelli, F. Reduction in antimicrobial resistance prevalence in Escherichia coli from a pig farm following withdrawal of group antimicrobial treatment. Vet. Microbiol. 2021, 258, 109125. [Google Scholar] [CrossRef] [PubMed]
  56. Mellor, K.C.; Petrovska, L.; Thomson, N.R.; Harris, K.; Reid, S.W.J.; Mather, A.E. Antimicrobial resistance diversity suggestive of distinct salmonella typhimurium sources or selective pressures in food-production animals. Front. Microbiol. 2019, 10, 708. [Google Scholar] [CrossRef] [PubMed]
  57. Tsigalou, C.; Konstantinidis, T.; Stavropoulou, E.; Bezirtzoglou, E.E.; Tsakris, A. Potential Elimination of Human Gut Resistome by Exploiting the Benefits of Functional Foods. Front. Microbiol. 2020, 11, 50. [Google Scholar] [CrossRef] [PubMed]
  58. Rafailidis, P.I.; Kofteridis, D. Proposed amendments regarding the definitions of multidrug-resistant and extensively drug-resistant bacteria. Expert Rev. Anti-Infect. Ther. 2021, 20, 139–146. [Google Scholar] [CrossRef]
  59. 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]
  60. Lai, C.K.C.; Ng, R.W.Y.; Leung, S.S.Y.; Hui, M.; Ip, M. Overcoming the rising incidence and evolving mechanisms of antibiotic resistance by novel drug delivery approaches—An overview. Adv. Drug Deliv. Rev. 2022, 181, 114078. [Google Scholar] [CrossRef]
  61. Li, L.; Xiao, Y.; Wang, C.; Olsen, R.H.; Meng, H.; Shi, L. Exploring the resistome, virulome, mobilome and microbiome along pork production chain using metagenomics. Int. J. Food Microbiol. 2022, 371, 109674. [Google Scholar] [CrossRef]
  62. Jans, C.; Sarno, E.; Collineau, L.; Meile, L.; Stärk, K.D.C.; Stephan, R. Consumer exposure to antimicrobial resistant bacteria from food at Swiss retail level. Front. Microbiol. 2018, 9, 362. [Google Scholar] [CrossRef]
  63. Jiang, Z.; Anwar, T.M.; Peng, X.; Biswas, S.; Elbediwi, M.; Li, Y.; Fang, W.; Yue, M. Prevalence and antimicrobial resistance of Salmonella recovered from pig-borne food products in Henan, China. Food Control 2021, 121, 107535. [Google Scholar] [CrossRef]
  64. Reid, C.J.; Blau, K.; Jechalke, S.; Smalla, K.; Djordjevic, S.P. Whole Genome Sequencing of Escherichia coli From Store-Bought Produce. Front. Microbiol. 2020, 10, 3050. [Google Scholar] [CrossRef]
  65. Kanokudom, S.; Assawakongkarat, T.; Akeda, Y.; Ratthawongjirakul, P.; Chuanchuen, R.; Chaichanawongsaroj, N. Rapid detection of extended spectrum β-lactamase producing Escherichia coli isolated from fresh pork meat and pig cecum samples using multiplex recombinase polymerase amplification and lateral flow strip analysis. PLoS ONE 2021, 16, e0248536. [Google Scholar] [CrossRef] [PubMed]
  66. Ferrocino, I.; Rantsiou, K.; McClure, R.; Kostic, T.; de Souza, R.S.C.; Lange, L.; FitzGerald, J.; Kriaa, A.; Cotter, P.; Maguin, E.; et al. The need for an integrated multi-OMICs approach in microbiome science in the food system. Compr. Rev. Food Sci. Food Saf. 2023, 22, 1082–1103. [Google Scholar] [CrossRef] [PubMed]
  67. Farm to Fork Strategy. Available online: https://food.ec.europa.eu/horizontal-topics/farm-fork-strategy_en (accessed on 6 June 2023).
  68. Pennone, V.; Cobo-Díaz, J.F.; Prieto, M.; Alvarez-Ordóñez, A. Application of genomics and metagenomics to improve food safety based on an enhanced characterisation of antimicrobial resistance. Curr. Opin. Food Sci. 2022, 43, 183–188. [Google Scholar] [CrossRef]
  69. Rocchetti, G.; Ferronato, G.; Sarv, V.; Kerner, K.; Venskutonis, P.R.; Lucini, L. Meat extenders from different sources as protein-rich alternatives to improve the technological properties and functional quality of meat products. Curr. Opin. Food Sci. 2023, 49, 100967. [Google Scholar] [CrossRef]
  70. Lu, N.; Ma, J.; Sun, D.W. Enhancing physical and chemical quality attributes of frozen meat and meat products: Mechanisms, techniques and applications. Trends Food Sci. Technol. 2022, 124, 63–85. [Google Scholar] [CrossRef]
  71. de Araújo, P.D.; Araújo, W.M.C.; Patarata, L.; Fraqueza, M.J. Understanding the main factors that influence consumer quality perception and attitude towards meat and processed meat products. Meat Sci. 2022, 193, 108952. [Google Scholar] [CrossRef]
  72. Van Reckem, E.; De Vuyst, L.; Weckx, S.; Leroy, F. Next-generation sequencing to enhance the taxonomic resolution of the microbiological analysis of meat and meat-derived products. Curr. Opin. Food Sci. 2021, 37, 58–65. [Google Scholar] [CrossRef]
  73. Chaudhary, P.; Kumar, Y. Recent advances in multiplex molecular techniques for meat species identification. J. Food Compos. Anal. 2022, 110, 104581. [Google Scholar] [CrossRef]
  74. World Consumption of Meat. Available online: https://www.theworldcounts.com/challenges/foods-and-beverages/world-consumption-of-meat (accessed on 6 June 2023).
  75. Brinck, J.E.; Lassen, S.B.; Forouzandeh, A.; Pan, T.; Wang, Y.-Z.; Monteiro, A.; Blavi, L.; Solà-Oriol, D.; Stein, H.H.; Su, J.-Q.; et al. Impacts of dietary copper on the swine gut microbiome and antibiotic resistome. Sci. Total Environ. 2023, 857, 159609. [Google Scholar] [CrossRef]
  76. Li, X.; Rensing, C.; Vestergaard, G.; Arumugam, M.; Nesme, J.; Gupta, S.; Brejnrod, A.D.; Sørensen, S.J. Metagenomic evidence for co-occurrence of antibiotic, biocide and metal resistance genes in pigs. Environ. Int. 2022, 158, 106899. [Google Scholar] [CrossRef]
  77. Luu, Q.H.; Nguyen, T.L.A.; Pham, T.N.; Vo, N.G.; Padungtod, P. Antimicrobial use in household, semi-industrialized, and industrialized pig and poultry farms in Viet Nam. Prev. Vet. Med. 2021, 189, 105292. [Google Scholar] [CrossRef] [PubMed]
  78. Pholwat, S.; Pongpan, T.; Chinli, R.; Rogawski 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] [PubMed]
  79. Sirichokchatchawan, W.; Apiwatsiri, P.; Pupa, P.; Saenkankam, I.; Khine, N.O.; Lekagul, A.; Lugsomya, K.; Hampson, D.J.; Prapasarakul, N. Reducing the Risk of Transmission of Critical Antimicrobial Resistance Determinants from Contaminated Pork Products to Humans in South-East Asia. Front. Microbiol. 2021, 12, 689015. [Google Scholar] [CrossRef]
  80. Wang, Y.; Sutton, N.B.; Zheng, Y.; Dong, H.; Rijnaarts, H.H.M. Seasonal variation in antibiotic resistance genes and bacterial phenotypes in swine wastewater during three-chamber anaerobic pond treatment. Environ. Res. 2023, 216, 114495. [Google Scholar] [CrossRef] [PubMed]
  81. Calero, G.C.; Gomez, N.C.; Benomar, N.; Montoro, B.P.; Knapp, C.W.; Galvez, A.; Abriouel, H. Deciphering Resistome and Virulome Diversity in a Porcine Slaughterhouse and Pork Products Through Its Production Chain. Front. Microbiol. 2018, 9, 2099. [Google Scholar] [CrossRef]
  82. Oswaldi, V.; Luth, S.; Dzierzon, J.; Meemken, D.; Schwarz, S.; Fessler, A.T.; Felix, B.; Langforth, S. Distribution and Characteristics of Listeria spp. in Pigs and Pork Production Chains in Germany. Microorganisms 2022, 10, 512. [Google Scholar] [CrossRef]
  83. Díaz-Jiménez, D.; García-Meniño, I.; Herrera, A.; Lestón, L.; Mora, A. Microbiological risk assessment of Turkey and chicken meat for consumer: Significant differences regarding multidrug resistance, mcr or presence of hybrid aEPEC/ExPEC pathotypes of E. coli. Food Control 2021, 123, 107713. [Google Scholar] [CrossRef]
  84. Das, T.; Paino, D.; Manoharan, A.; Farrell, J.; Whiteley, G.; Kriel, F.H.; Glasbey, T.; Manos, J. Conditions under which glutathione disrupts the biofilms and improves antibiotic efficacy of both ESKAPE and NON-ESKAPE species. Front. Microbiol. 2019, 10, 2000. [Google Scholar] [CrossRef]
  85. Zohra, T.; Numan, M.; Ikram, A.; Salman, M.; Khan, T.; Din, M.; Salman, M.; Farooq, A.; Amir, A.; Ali, M. Cracking the Challenge of Antimicrobial Drug Resistance with CRISPR/Cas9, Nanotechnology and Other Strategies in ESKAPE Pathogens. Microorganisms 2021, 9, 954. [Google Scholar] [CrossRef]
  86. Klimienė, I.; Virgailis, M.; Kerzienė, S.; Šiugždinienė, R.; Mockeliūnas, R.; Ružauskas, M. Evaluation of genotypical antimicrobial resistance in ESBL producing Escherichia coli phylogenetic groups isolated from retail poultry meat. J. Food Saf. 2018, 38, e12370. [Google Scholar] [CrossRef]
  87. Correia Santos, S.; Fraqueza, M.J.; Elias, M.; Salvador Barreto, A.; Semedo-Lemsaddek, T. Traditional dry smoked fermented meat sausages: Characterization of autochthonous enterococci. LWT-Food Sci. Technol. 2017, 79, 410–415. [Google Scholar] [CrossRef]
  88. Yu, T.; Jiang, X.; Liang, Y.; Zhu, Y.; Tian, J.; Ying, H.; Wang, X.; Shi, L. Characterization and horizontal transfer of antimicrobial resistance genes and integrons in bacteria isolated from cooked meat products in China. J. Food Prot. 2017, 80, 2048–2055. [Google Scholar] [CrossRef] [PubMed]
  89. Bouchami, O.; Fraqueza, M.J.; Faria, N.A.; Alves, V.; Lawal, O.U.; de Lencastre, H.; Miragaia, M. Evidence for the dissemination to humans of methicillin-resistant Staphylococcus aureus ST398 through the pork production chain: A study in a portuguese slaughterhouse. Microorganisms 2020, 8, 1892. [Google Scholar] [CrossRef] [PubMed]
  90. Cao, H.; Bougouffa, S.; Park, T.-J.; Lau, A.; Tong, M.-K.; Chow, K.-H.; Ho, P.-L. Sharing of Antimicrobial Resistance Genes between Humans and Food Animals. mSystems 2022, 7, e00775-22. [Google Scholar] [CrossRef]
  91. Lauteri, C.; Festino, A.R.; Conter, M.; Vergara, A. Prevalence and antimicrobial resistance profile in Salmonella spp. isolates from swine food chain. Ital. J. Food Saf. 2022, 11, 9980. [Google Scholar] [CrossRef]
  92. Liu, X.; Li, R.; Chan, E.W.-C.; Chen, S. Complete Genetic Analysis of Plasmids Carrying Multiple Resistance, Virulence, and Phage-Like Genes in Foodborne Escherichia coli Isolate. Microbiol. Spectr. 2023, 11, e02820-22. [Google Scholar] [CrossRef]
  93. Sin, M.; Yoon, S.; Kim, Y.B.; Noh, E.B.; Seo, K.W.; Lee, Y.J. Molecular characteristics of antimicrobial resistance determinants and integrons in Salmonella isolated from chicken meat in Korea. J. Appl. Poult. Res. 2020, 29, 502–514. [Google Scholar] [CrossRef]
  94. Wang, W.; Chen, J.; Shao, X.; Huang, P.; Zha, J.; Ye, Y. Occurrence and antimicrobial resistance of Salmonella isolated from retail meats in Anhui, China. Food Sci. Nutr. 2021, 9, 4701–4710. [Google Scholar] [CrossRef]
  95. Wang, Y.T.; Lin, Y.T.; Wan, T.W.; Wang, D.Y.; Lin, H.Y.; Lin, C.Y.; Chen, Y.C.; Teng, L.J. Distribution of antibiotic resistance genes among Staphylococcus species isolated from ready-to-eat foods. J. Food Drug Anal. 2019, 27, 841–848. [Google Scholar] [CrossRef]
  96. Guerrero-Ramos, E.; Molina-González, D.; Blanco-Morán, S.; Igrejas, G.; Poeta, P.; Alonso-Calleja, C.; Capita, R. Prevalence, antimicrobial resistance, and genotypic characterization of vancomycin-resistant enterococci in meat preparations. J. Food Prot. 2016, 79, 748–756. [Google Scholar] [CrossRef]
  97. Belloso Daza, M.V.; Milani, G.; Cortimiglia, C.; Pietta, E.; Bassi, D.; Cocconcelli, P.S. Genomic Insights of Enterococcus faecium UC7251, a Multi-Drug Resistant Strain from Ready-to-Eat Food, Highlight the Risk of Antimicrobial Resistance in the Food Chain. Front. Microbiol. 2022, 13, 894241. [Google Scholar] [CrossRef] [PubMed]
  98. Zhang, L.; Fu, Y.; Xiong, Z.; Ma, Y.; Wei, Y.; Qu, X.; Zhang, H.; Zhang, J.; Liao, M. Highly Prevalent Multidrug-Resistant Salmonella from Chicken and Pork Meat at Retail Markets in Guangdong, China. Front. Microbiol. 2018, 9, 2104. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, J.; Wei, X.; Fan, M. Assessment of Antibiotic Susceptibility within Lactic Acid Bacteria and Coagulase-Negative Staphylococci Isolated from Hunan Smoked Pork, a Naturally Fermented Meat Product in China. J. Food Sci. 2018, 83, 1707–1715. [Google Scholar] [CrossRef] [PubMed]
  100. Heo, E.J.; Ko, E.K.; Kang, H.J.; Kim, Y.J.; Park, H.J.; Wee, S.H.; Moon, J.S. Prevalence and Antimicrobial Characteristics of Shiga Toxin-Producing Escherichia coli Isolates from Pork in Korea. Foodborne Pathog. Dis. 2020, 17, 602–607. [Google Scholar] [CrossRef]
  101. Li, L.; Heidemann Olsen, R.; Ye, L.; Yan, H.; Nie, Q.; Meng, H.; Shi, L. Antimicrobial Resistance and Resistance Genes in Aerobic Bacteria Isolated from Pork at Slaughter. J. Food Prot. 2016, 79, 589–597. [Google Scholar] [CrossRef]
  102. Liu, Z.; Klümper, U.; Shi, L.; Ye, L.; Li, M. From pig breeding environment to subsequently produced pork: Comparative analysis of antibiotic resistance genes and bacterial community composition. Front. Microbiol. 2019, 10, 43. [Google Scholar] [CrossRef]
  103. Li, L.; Ye, L.; Yu, L.; Zhou, C.; Meng, H. Characterization of Extended Spectrum Beta-Lactamase Producing Enterobacteria and Methicillin-Resistant Staphylococcus aureus Isolated from Raw Pork and Cooked Pork Products in South China. J. Food Sci. 2016, 81, M1773–M1777. [Google Scholar] [CrossRef]
  104. Bacci, C.; Lanzoni, E.; Vismarra, A.; Alpigiani, I.; Nuvoloni, R.; Bonardi, S.; Brindani, F. Antibiotic resistance and resistance genes in Salmonella enterica isolated from pork meat and pig carcasses in Northern Italy. Large Anim. Rev. 2014, 20, 201–207. [Google Scholar]
  105. Chajecka-Wierzchowska, W.; Zadernowska, A.; Nalepa, B.; Sierpinska, M.; Laniewska-Trokenheim, L. Coagulase-negative staphylococci (CoNS) isolated from ready-to-eat food of animal origin—Phenotypic and genotypic antibiotic resistance. Food Microbiol. 2015, 46, 222–226. [Google Scholar] [CrossRef]
  106. Lawal, O.U.; Fraqueza, M.J.; Bouchami, O.; Worning, P.; Bartels, M.D.; Gonçalves, M.L.; Paixao, P.; Gonçalves, E.; Toscano, C.; Empel, J.; et al. Foodborne origin and local and global spread of staphylococcus saprophyticus causing human urinary tract infections. Emerg. Infect. Dis. 2021, 27, 880–893. [Google Scholar] [CrossRef]
  107. Henriques, A.R.; Melo Cristino, J.; Fraqueza, M.J. Genetic characterization of listeria monocytogenes isolates from industrial and retail ready-to-eat meat-based foods and their relationship with clinical strains from human listeriosis in Portugal. J. Food Prot. 2017, 80, 551–560. [Google Scholar] [CrossRef] [PubMed]
  108. Rolain, J.M. Food and human gut as reservoirs of transferable antibiotic resistance encoding genes. Front. Microbiol. 2013, 4, 173. [Google Scholar] [CrossRef] [PubMed]
  109. Stanton, I.C.; Bethel, A.; Leonard, A.F.C.; Gaze, W.H.; Garside, R. Existing evidence on antibiotic resistance exposure and transmission to humans from the environment: A systematic map. Environ. Evid. 2022, 11, 8. [Google Scholar] [CrossRef] [PubMed]
  110. Igrejas, G.; Correia, S.; Silva, V.; Hébraud, M.; Caniça, M.; Torres, C.; Gomes, C.; Nogueira, F.; Poeta, P. Planning a one health case study to evaluate Methicillin resistant Staphylococcus aureus and its economic burden in Portugal. Front. Microbiol. 2018, 9, 2964. [Google Scholar] [CrossRef]
  111. Neyaz, L.; Rajagopal, N.; Wells, H.; Fakhr, M.K. Molecular Characterization of Staphylococcus aureus Plasmids Associated with Strains Isolated From Various Retail Meats. Front. Microbiol. 2020, 11, 223. [Google Scholar] [CrossRef]
  112. Okorie-Kanu, O.J.; Anyanwu, M.U.; Ezenduka, E.V.; Mgbeahuruike, A.C.; Thapaliya, D.; Gerbig, G.; Ugwuijem, E.E.; Okorie-Kanu, C.O.; Agbowo, P.; Olorunleke, S.; et al. Molecular epidemiology, genetic diversity and antimicrobial resistance of Staphylococcus aureus isolated from chicken and pig carcasses, and carcass handlers. PLoS ONE 2020, 15, e0232913. [Google Scholar] [CrossRef]
  113. Şanlıbaba, P. Prevalence, antibiotic resistance, and enterotoxin production of Staphylococcus aureus isolated from retail raw beef, sheep, and lamb meat in Turkey. Int. J. Food Microbiol. 2022, 361, 109461. [Google Scholar] [CrossRef]
  114. Barros, E.M.; Martin, M.J.; Selleck, E.M.; Lebreton, F.; Sampaio, J.L.M.; Gilmore, M.S. Daptomycin Resistance and Tolerance Due to Loss of Function in Staphylococcus aureus dsp1 and asp23. Antimicrob. Agents Chemother. 2019, 63, e01542-18. [Google Scholar] [CrossRef]
  115. Zhang, J.; Xu, J.; Lei, H.; Liang, H.; Li, X.; Li, B. The development of variation-based rifampicin resistance in Staphylococcus aureus deciphered through genomic and transcriptomic study. J. Hazard. Mater. 2023, 442, 130112. [Google Scholar] [CrossRef]
  116. Fraqueza, M.J.; Laranjo, M.; Elias, M.; Patarata, L. Microbiological hazards associated with salt and nitrite reduction in cured meat products: Control strategies based on antimicrobial effect of natural ingredients and protective microbiota. Curr. Opin. Food Sci. 2021, 38, 32–39. [Google Scholar] [CrossRef]
  117. Vestergaard, M.; Nøhr-Meldgaard, K.; Ingmer, H. Multiple pathways towards reduced membrane potential and concomitant reduction in aminoglycoside susceptibility in Staphylococcus aureus. Int. J. Antimicrob. Agents 2018, 51, 132–135. [Google Scholar] [CrossRef]
  118. Correia, S.; Silva, V.; García-Díez, J.; Teixeira, P.; Pimenta, K.; Pereira, J.E.; Oliveira, S.; Rocha, J.; Manaia, C.M.; Igrejas, G.; et al. One Health Approach Reveals the Absence of Methicillin-Resistant Staphylococcus aureus in Autochthonous Cattle and Their Environments. Front. Microbiol. 2019, 10, 2735. [Google Scholar] [CrossRef]
  119. Hau, S.J.; Haan, J.S.; Davies, P.R.; Frana, T.; Nicholson, T.L. Antimicrobial resistance distribution differs among methicillin resistant Staphylococcus aureus sequence type (ST) 5 isolates from health care and agricultural sources. Front. Microbiol. 2018, 9, 2102. [Google Scholar] [CrossRef] [PubMed]
  120. Watkins, R.R.; Holubar, M.; David, M.Z. Antimicrobial Resistance in Methicillin-Resistant Staphylococcus aureus to Newer Antimicrobial Agents. Antimicrob. Agents Chemother. 2019, 63, e01216-19. [Google Scholar] [CrossRef] [PubMed]
  121. Zhang, H.; Qin, L.; Jin, C.; Ju, H.; Jiang, R.; Li, L.; Zhang, H.; Gao, W.; Wei, X.; Dong, H.; et al. Molecular Characteristics and Antibiotic Resistance of Staphylococcus aureus Isolated from Patient and Food Samples in Shijiazhuang, China. Pathogens 2022, 11, 1333. [Google Scholar] [CrossRef] [PubMed]
  122. Lv, G.; Jiang, R.; Zhang, H.; Wang, L.; Li, L.; Gao, W.; Zhang, H.; Pei, Y.; Wei, X.; Dong, H.; et al. Molecular Characteristics of Staphylococcus aureus From Food Samples and Food Poisoning Outbreaks in Shijiazhuang, China. Front. Microbiol. 2021, 12, 652276. [Google Scholar] [CrossRef]
  123. Bonardi, S.; Cabassi, C.S.; Manfreda, G.; Parisi, A.; Fiaccadori, E.; Sabatino, A.; Cavirani, S.; Bacci, C.; Rega, M.; Spadini, C.; et al. Survey on Carbapenem-Resistant Bacteria in Pigs at Slaughter and Comparison with Human Clinical Isolates in Italy. Antibiotics 2022, 11, 777. [Google Scholar] [CrossRef]
  124. Moosavian, M.; Emam, N. The first report of emerging mobilized colistin-resistance (Mcr) genes and ERIC-PCR typing in Escherichia coli and Klebsiella pneumoniae clinical isolates in southwest Iran. Infect. Drug Resist. 2019, 12, 1001–1010. [Google Scholar] [CrossRef]
  125. Teklu, D.S.; Negeri, A.A.; Legese, M.H.; Bedada, T.L.; Woldemariam, H.K.; Tullu, K.D. Extended-spectrum beta-lactamase production and multi-drug resistance among Enterobacteriaceae isolated in Addis Ababa, Ethiopia. Antimicrob. Resist. Infect. Control 2019, 8, 39. [Google Scholar] [CrossRef]
  126. Alizadeh, N.; Rezaee, M.A.; Kafil, H.S.; Hasani, A.; Barhaghi, M.H.S.; Milani, M.; Sefidan, F.Y.; Memar, M.Y.; Lalehzadeh, A.; Ghotaslo, R. Evaluation of Resistance Mechanisms in Carbapenem-Resistant Enterobacteriaceae. Infect. Drug Resist. 2020, 13, 1377–1385. [Google Scholar] [CrossRef]
  127. Castanheira, M.; Deshpande, L.M.; Mendes, R.E.; Canton, R.; Sader, H.S.; Jones, R.N. Variations in the Occurrence of Resistance Phenotypes and Carbapenemase Genes Among Enterobacteriaceae Isolates in 20 Years of the SENTRY Antimicrobial Surveillance Program. Open Forum Infect. Dis. 2019, 6, S23–S33. [Google Scholar] [CrossRef] [PubMed]
  128. 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]
  129. WHO Publishes List of Bacteria for Which New Antibiotics Are Urgently Needed. Available online: https://www.who.int/news/item/27-02-2017-who-publishes-list-of-bacteria-for-which-new-antibiotics-are-urgently-needed (accessed on 6 June 2023).
  130. Jiang, B.; Du, P.; Jia, P.; Liu, E.; Kudinha, T.; Zhang, H.; Li, D.; Xu, Y.; Xie, L.; Yang, Q. Antimicrobial Susceptibility and Virulence of mcr-1-Positive Enterobacteriaceae in China, a Multicenter Longitudinal Epidemiological Study. Front. Microbiol. 2020, 11, 1611. [Google Scholar] [CrossRef] [PubMed]
  131. Lombardi, G.; Tanzarella, E.S.; Cutuli, S.L.; De Pascale, G. Treatment of severe infections caused by ESBL or carbapenemases-producing Enterobacteriaceae. Med. Intensiv. Engl. Ed. 2022, 47, 34–44. [Google Scholar] [CrossRef]
  132. Richter, L.; du Plessis, E.M.; Duvenage, S.; Korsten, L. Occurrence, Phenotypic and Molecular Characterization of Extended-Spectrum- and AmpC- β-Lactamase Producing Enterobacteriaceae Isolated from Selected Commercial Spinach Supply Chains in South Africa. Front. Microbiol. 2019, 11, 638. [Google Scholar] [CrossRef]
  133. Ibrahim, M.E.; Abbas, M.; Al-Shahrai, A.M.; Elamin, B.K. Phenotypic Characterization and Antibiotic Resistance Patterns of Extended-Spectrum β-Lactamase- and AmpC β-Lactamase-Producing Gram-Negative Bacteria in a Referral Hospital, Saudi Arabia. Can. J. Infect. Dis. Med. Microbiol. 2019, 2019, 6054694. [Google Scholar] [CrossRef]
  134. Rubee Chanu, T.; K Shah, P.; Soni, S.; Ghosh, A. Phenotypic detection of extended spectrum, AmpC, Metallo beta-lactamases and their coexistence in clinical isolates of commonly isolated gram negativebacteria in GKGH hospital, Bhuj. IP Int. J. Med. Microbiol. Trop. Dis. 2019, 5, 52–56. [Google Scholar] [CrossRef]
  135. Ejikeugwu, C.; Nworie, O.; Saki, M.; Al-Dahmoshi, H.O.M.; Al-Khafaji, N.S.K.; Ezeador, C.; Nwakaeze, E.; Eze, P.; Oni, E.; Obi, C.; et al. Metallo-β-lactamase and AmpC genes in Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa isolates from abattoir and poultry origin in Nigeria. BMC Microbiol. 2021, 21, 124. [Google Scholar] [CrossRef]
  136. Sultan, I.; Siddiqui, M.T.; Gogry, F.A.; Haq, Q.M.R. Molecular characterization of resistance determinants and mobile genetic elements of ESBL producing multidrug-resistant bacteria from freshwater lakes in Kashmir, India. Sci. Total Environ. 2022, 827, 154221. [Google Scholar] [CrossRef]
  137. Carlet, J.; Jarlier, V.; Acar, J.; Debaere, O.; Dehaumont, P.; Grandbastien, B.; Le Coz, P.; Lina, G.; Pean, Y.; Rambaud, C.; et al. Trends in Antibiotic Consumption and Resistance in France over 20 Years: Large and Continuous Efforts but Contrasting Results. Open Forum Infect. Dis. 2020, 7, ofaa452. [Google Scholar] [CrossRef]
  138. Seenama, C.; Thamlikitkul, V.; Ratthawongjirakul, P. Multilocus sequence typing and bla (ESBL) characterization of extended-spectrum beta-lactamase-producing Escherichia coli isolated from healthy humans and swine in Northern Thailand. Infect. Drug Resist. 2019, 12, 2201–2214. [Google Scholar] [CrossRef] [PubMed]
  139. Nguyen, M.N.; Hoang, H.T.T.; Xavier, B.B.; Lammens, C.; Le, H.T.; Hoang, N.T.B.; Nguyen, S.T.; Pham, N.T.; Goossens, H.; Dang, A.D.; et al. Prospective One Health genetic surveillance in Vietnam identifies distinct bla(CTX-M)-harbouring Escherichia coli in food-chain and human-derived samples. Clin. Microbiol. Infect. 2021, 27, 1515.e1–1515.e8. [Google Scholar] [CrossRef] [PubMed]
  140. Ayala, D.I.; Cook, P.W.; Franco, J.G.; Bugarel, M.; Kottapalli, K.R.; Loneragan, G.H.; Brashears, M.M.; Nightingale, K.K. A systematic approach to identify and characterize the effectiveness and safety of novel probiotic strains to control foodborne pathogens. Front. Microbiol. 2019, 10, 1108. [Google Scholar] [CrossRef] [PubMed]
  141. Markwart, R.; Willrich, N.; Haller, S.; Noll, I.; Koppe, U.; Werner, G.; Eckmanns, T.; Reuss, A. The rise in vancomycin-resistant Enterococcus faecium in Germany: Data from the German Antimicrobial Resistance Surveillance (ARS). Antimicrob. Resist. Infect. Control 2019, 8, 147. [Google Scholar] [CrossRef]
  142. Chilambi, G.S.; Hinks, J.; Matysik, A.; Zhu, X.; Choo, P.Y.; Liu, X.; Chan-Park, M.B.; Bazan, G.C.; Kline, K.A.; Rice, S.A. Enterococcus faecalis Adapts to Antimicrobial Conjugated Oligoelectrolytes by Lipid Rearrangement and Differential Expression of Membrane Stress Response Genes. Front. Microbiol. 2020, 11, 155. [Google Scholar] [CrossRef]
  143. Di Cesare, A.; Frangipani, E.; Citterio, B.; Sabatino, R.; Corno, G.; Fontaneto, D.; Mangiaterra, G.; Bencardino, D.; Zoppi, S.; Di Blasio, A.; et al. Class 1 integron and Enterococcus spp. abundances in swine farms from the “Suckling piglets” to the “Fatteners” production category. Vet. Microbiol. 2022, 274, 109576. [Google Scholar] [CrossRef]
  144. Liu, M.; Kemper, N.; Volkmann, N.; Schulz, J. Resistance of enterococcus spp. in dust from farm animal houses: A retrospective study. Front. Microbiol. 2018, 9, 3074. [Google Scholar] [CrossRef]
  145. Mahony, A.A.; Buultjens, A.H.; Ballard, S.A.; Grabsch, E.A.; Xie, S.; Seemann, T.; Stuart, R.L.; Kotsanas, D.; Cheng, A.; Heffernan, H.; et al. Vancomycin-resistant Enterococcus faecium sequence type 796-rapid international dissemination of a new epidemic clone. Antimicrob. Resist. Infect. Control 2018, 7, 44. [Google Scholar] [CrossRef]
  146. Abbo, L.; Shukla, B.S.; Giles, A.; Aragon, L.; Jimenez, A.; Camargo, J.F.; Simkins, J.; Sposato, K.; Tran, T.T.; Diaz, L.; et al. Linezolid-and vancomycin-resistant Enterococcus faecium in solid organ transplant recipients: Infection control and antimicrobial stewardship using whole genome sequencing. Clin. Infect. Dis. 2019, 69, 259–265. [Google Scholar] [CrossRef]
  147. Farman, M.; Yasir, M.; Al-Hindi, R.R.; Farraj, S.A.; Jiman-Fatani, A.A.; Alawi, M.; Azhar, E.I. Genomic analysis of multidrug-resistant clinical Enterococcus faecalis isolates for antimicrobial resistance genes and virulence factors from the western region of Saudi Arabia. Antimicrob. Resist. Infect. Control 2019, 8, 55. [Google Scholar] [CrossRef]
  148. Kang, X.; Wei, Y.; Fan, X.; Luo, S.; Luo, X.; Zhao, S.; Wang, G. Analysis of virulence genes, drug resistance detection, and pathogenicity in Enterococcus from farm animals. Microb. Pathog. 2022, 171, 105745. [Google Scholar] [CrossRef] [PubMed]
  149. Deshpande, L.M.; Castanheira, M.; Flamm, R.K.; Mendes, R.E. Evolving oxazolidinone resistance mechanisms in a worldwide collection of enterococcal clinical isolates: Results from the SENTRY Antimicrobial Surveillance Program. J. Antimicrob. Chemother. 2018, 73, 2314–2322. [Google Scholar] [CrossRef] [PubMed]
  150. Sacramento, A.G.; Andrade, A.C.D.; Teotonio, B.N.; de Oliveira Santos, L.M.; da Silva, L.C.B.A.; Lincopan, N.; Sellera, F.P. WHO critical priority van-type vancomycin-resistant Enterococcus in dogs and cats. Prev. Vet. Med. 2022, 202, 105614. [Google Scholar] [CrossRef] [PubMed]
  151. Bender, J.K.; Cattoir, V.; Hegstad, K.; Sadowy, E.; Coque, T.M.; Westh, H.; Hammerum, A.M.; Schaffer, K.; Burns, K.; Murchan, S.; et al. Update on prevalence and mechanisms of resistance to linezolid, tigecycline and daptomycin in enterococci in Europe: Towards a common nomenclature. Drug Resist. Updates 2018, 40, 25–39. [Google Scholar] [CrossRef]
  152. Larsen, J.; Schonheyder, H.C.; Lester, C.H.; Olsen, S.S.; Porsbo, L.J.; Garcia-Migura, L.; Jensen, L.B.; Bisgaard, M.; Hammerum, A.M. Porcine-origin gentamicin-resistant Enterococcus faecalis in humans, Denmark. Emerg. Infect. Dis. 2010, 16, 682–684. [Google Scholar] [CrossRef]
  153. Denissen, J.; Reyneke, B.; Waso-Reyneke, M.; Havenga, B.; Barnard, T.; Khan, S.; Khan, W. Prevalence of ESKAPE pathogens in the environment: Antibiotic resistance status, community-acquired infection and risk to human health. Int. J. Hyg. Environ. Health 2022, 244, 114006. [Google Scholar] [CrossRef]
  154. Ma, Y.X.; Wang, C.Y.; Li, Y.Y.; Li, J.; Wan, Q.Q.; Chen, J.H.; Tay, F.R.; Niu, L.N. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. Adv. Sci. 2020, 7, 1901872. [Google Scholar] [CrossRef]
  155. de Aledo, M.G.; González-Bardanca, M.; Blasco, L.; Pacios, O.; Bleriot, I.; Fernández-García, L.; Fernández-Quejo, M.; López, M.; Bou, G.; Tomás, M. Crispr-cas, a revolution in the treatment and study of eskape infections: Pre-clinical studies. Antibiotics 2021, 10, 756. [Google Scholar] [CrossRef]
  156. Intra, J.; Carcione, D.; Sala, R.M.; Siracusa, C.; Brambilla, P.; Leoni, V. Antimicrobial Resistance Patterns of Enterobacter cloacae and Klebsiella aerogenes Strains Isolated from Clinical Specimens: A Twenty-Year Surveillance Study. Antibiotics 2023, 12, 775. [Google Scholar] [CrossRef]
Figure 1. Main mechanisms of antimicrobial activity.
Figure 1. Main mechanisms of antimicrobial activity.
Microorganisms 11 02581 g001
Figure 2. Horizontal gene transfer through conjugation. (a) Microbial community with antimicrobial susceptible bacteria (green) and antimicrobial resistant bacteria (blue); (b) Fusion between resistant and susceptible bacteria, allowing the transfer of genetic information through conjugation pili; (c) Microbial community with antimicrobial resistant bacteria.
Figure 2. Horizontal gene transfer through conjugation. (a) Microbial community with antimicrobial susceptible bacteria (green) and antimicrobial resistant bacteria (blue); (b) Fusion between resistant and susceptible bacteria, allowing the transfer of genetic information through conjugation pili; (c) Microbial community with antimicrobial resistant bacteria.
Microorganisms 11 02581 g002
Figure 3. Biochemical mechanisms involved in the different bacterial resistance processes.
Figure 3. Biochemical mechanisms involved in the different bacterial resistance processes.
Microorganisms 11 02581 g003
Table 1. Antimicrobial resistance genes identified in different types of meat and meat products.
Table 1. Antimicrobial resistance genes identified in different types of meat and meat products.
Type of FoodMicrobiotaAntimicrobial Resistance GenesReference
Raw poultry, pork and beefEnterococcus spp.Vancomycin: vanA, vanB and vanC1,2,3[96]
Tetracycline: tetM, tetL
Erythromycin: ermA and ermB
Quinupristin-dalfopristin: vat[D] and vat[E]
Retail poultry meatEscherichia coliβ-lactam: blaTEM, blaSHV, blaCMY-2 and blaCTX-M[86]
Sulphamethoxazole: sul2
Tetracycline: tetA and tetB
Chloramphenicol: cmlA
Aminoglycoside: aphA1 and aadA
Trimethoprim: dfrA1
Bull-cooked meat products Enterobacter spp.
Escherichia coli
Citrobacter spp.
Pseudomonas spp.
β-lactam: blaTEM-1 and blaCTX-M-14[88]
Gentamicin: aac(3)-IIa
Streptomycin: strA and strB
Quinolone: qnrB and qnrS
Sulphamethoxazole: sul1, sul2 and sul3
Chloramphenicol: cat1 and cat3
Tetracycline: tetM. tetA and tetB
Animal-based products (ready-to-eat food)Staphylococcus saprophyticus
Staphylococcus sciuri
Staphylococcus xylosus.
Oxacillin: mecA[95]
β-lactam: blaZ
Tetracycline: tetK
Erythromycin: msrA, msrB, ermA
Gentamycin: aacA-aphD
Fusidic acid: fusD
Trimethoprim/sulfamethoxazole: dfrG
Chicken meatSalmonella Albany
Salmonella Virchow
Salmonella Enteritidis
Salmonella Infantis
β-lactam: blaCTZ-M-15, blaCTX-M-79 and blaCMY-2[93]
Tetracycline: tetA and tetB
Sulfonamide: sul1 and sul2
Chloramphenicol: catA1 and cmlA
Retail meat (pork, chicken and duck)Salmonella Enteritidis
Salmonella Typhimurium
Salmonella Typhi
Salmonella Goldcoast
Salmonella Ouakam
Salmonella Paratyphi
Tetracycline: tetA[94]
β-lactam: blaTEM
Aminoglycoside: aadA1 and aadA2
Sulfonamide: sul1 and sul2
Dry fermented Italian salamiEnterococcus faecium UC7251Ampicillin: pbp5-S1/R20[97]
Gentamycin: aac(6′)-li
Kanamycin: aph(3′)-lll
Streptomycin: aad6 and aadE
Erythromycin: ermB, mrsC and sat4
Clindamycin: ermB, lnuB and lsaE
Tylosine: ermB
Tetracycline: tetL and tetM
Traditional pork dry sausagesSalmonella Enteritidis
Salmonella Typhi
Salmonella Typhimurium
Quinolone: gyrA and parC[91]
Chloramphenicol: catA1
Trimethoprim: drfA
Tetracycline: tetA and tetB
Nitrofurantoin: nfsA and nfsB
Ampicillin: blaTEM
Chicken meatEscherichia coli isolate 1108β-lactam: blaNDM-1, blaTEM-1, blaCTZX-M-64 and blaCMY-2[92]
Bleomycin: bleMBL
Sulfonamide: sul1 and sul2
Tetracycline: tetA and tetR
Aminoglycosides: strA
Quinolone: oqxA and oqxB
Phenicol: floR
Streptomycin: aadA2
Trimethoprim: dfrA12
Retail meat (chicken and pork)Salmonella Kentucky
Salmonella Indiana
Salmonella Derby
Salmonella Typhimurium
Salmonella Litchfield
Salmonella Schwarzengrun
β-lactam: blaCTX-M-55, blaTEM-206, blaTEM-214, blaOXA-1, blaCTX-M-123, blaTEM-1, blaCTX-M-64 and blaCTX-M-15[98]
Naturally fermented smoked porkStaphylococcus carnosus
Lactobacillus plantarum
Labctobacillus brevis
Lactobacillus sakei
Weissella confusa
Weissella cibaria
Tetracycline: tetO and tetM
Erythromycin: ereA
Chloramphenicol: catA
Streptomycin: strA and strB
[99]
Pork meatAeromonas aquariorum
Aeromonas hydrophila
Aeromonas jandaei
Aeromonas veronii
Acinetobacter baumannii
Acinetobacter bereziniae
Acinetobacter johnsonii
Acinetobacter septicus
Acinetobacter ursingii
Citrobacter sp.
Citrobacter freundii
Citrobacter murliniae
Enterobacteriaceae
Enterobacter sp.
Enterobacter asburiae
Enterobacter cloacae
Enterobacter hormaechei
Enterobacter ludwigii
Escherichia coli
Klebsiella sp.
Klebsiella oxytoca
Klebsiella terrigena
Lactobacillus casei
Leclercia sp.
Lactococcus garvieae
Lactococcus lactis
Micrococcus caseolyticus
Myroides phaeus
Myroides marinus
Myroides odoratimimus
Oceanobacillus
Pantoea sp.
Pantoea dispersa
Pantoea agglomerans
Proteus penneri
Providencia alcalifaciens
Pseudomonas sp.
Raoultella sp.
Raoultella terrigena
Serratia sp.
Serratia marcescens
Sphingobacterium
Staphylococcus sp.
Staphylococcus sciuri
Staphylococcus epidermidis
Vibrio cincinnatiensis
Wautersiella falsenii genomovar 1
Kurthia sp.
Bacillus sp.
Morganella sp.
Micrococcus caseolyticus
Vagococcus sp.
Raoultella ornithinolytica
Comamonas sp.
Budvicia sp.
Aeromonas sp.
Klebsiella sp.
β-lactam: blaTEM, blaCTX-M, blaCMY-2
Tetracycline: tetA, tetC, tetE, tetK, tetL, tetM and tetS
Sulfonamide: sul1 and sul2
Aminoglycoside: aadA and aphA-1
Chloramphenicol: cmlA
Macrolide: ermB
Florfenicol: floR
[100,101,102]
Raw and cooked porkCitrobacter freundii
Serratia marcescens
Escherichia coli
β-lactams: blaTEM, blaCTX-M-1, blaSHV and blaCTX-M-9 [103]
Pork meat and pork meat preparations (cotechino, hamburger, sausages and Zuccotto of Bismantova)Salmonella Derby
Salmonella Typhimurium
Salmonella Bredeney
Salmonella London
Salmonella Anatum
Salmonella Agona
Salmonella Virchow
Salmonella Senftenberg
Salmonella Livingstone
Salmonella India
Salmonella Heidelberg
Salmonella Bovis-morbificans
Salmonella Coeln
Ampicillin: blaPSE-1
Gentamicin: ant (2″)-Ia
Sulfamethoxazole: sul1
Tetracycline: tetA, tetB, tetG and marRAB
[104]
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

Conceição, S.; Queiroga, M.C.; Laranjo, M. Antimicrobial Resistance in Bacteria from Meat and Meat Products: A One Health Perspective. Microorganisms 2023, 11, 2581. https://doi.org/10.3390/microorganisms11102581

AMA Style

Conceição S, Queiroga MC, Laranjo M. Antimicrobial Resistance in Bacteria from Meat and Meat Products: A One Health Perspective. Microorganisms. 2023; 11(10):2581. https://doi.org/10.3390/microorganisms11102581

Chicago/Turabian Style

Conceição, Sara, Maria Cristina Queiroga, and Marta Laranjo. 2023. "Antimicrobial Resistance in Bacteria from Meat and Meat Products: A One Health Perspective" Microorganisms 11, no. 10: 2581. https://doi.org/10.3390/microorganisms11102581

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