Next Article in Journal
Mapping Selected Emergent Marine Toxin-Producing Organisms Using Historical Samples with Two Methods (Biosensors and Real-Time PCR): A Comparison of Resolution
Previous Article in Journal
Culture-Independent Quantification of Legionella pneumophila in Evaporative Cooling Systems Using Immunomagnetic Separation Coupled with Flow Cytometry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Impact of In-Water vs. In-Feed Chlortetracycline and Tiamulin Administration in Piglets on the Fecal Prevalence and Antimicrobial Resistance of Salmonella

1
Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
2
Department of Animal Sciences & Industry, College of Agriculture, Kansas State University, Manhattan, KS 66506, USA
3
Department of Statistics, College of Arts and Sciences, Kansas State University, Manhattan, KS 66506, USA
4
Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
*
Author to whom correspondence should be addressed.
Appl. Microbiol. 2024, 4(1), 297-311; https://doi.org/10.3390/applmicrobiol4010020
Submission received: 4 January 2024 / Revised: 24 January 2024 / Accepted: 26 January 2024 / Published: 28 January 2024

Abstract

:
Antimicrobial Resistance (AMR) in bacteria is a growing public health concern in the US and around the world threatening the continual use of antimicrobials. In pigs, the oral route, either in-feed or in-water, is by far the most common route of administration of antimicrobials. Because the distribution of the antibiotic in the gut and the dosages are different, the impact of in-feed vs. in-water administration of antibiotics on the prevalence of pathogens, such as Salmonella, and the development of AMR are likely to be different. Therefore, a study was conducted to compare in-feed vs. in-water administrations of chlortetracycline (CTC) and/or tiamulin on the fecal prevalence and AMR profiles of Salmonella in nursery piglets. A total of 1296 weaned piglets, housed in 48 pens (27 piglets per pen), were assigned randomly to six treatment groups: Control (no antibiotic), in-feed CTC, in-water CTC, in-feed tiamulin, in-water tiamulin, or in-feed CTC and tiamulin. Fecal samples (n = 1440) were collected randomly from five piglets from each pen during the pre-treatment (days 7, 0), treatment (days 7, 14), and post-treatment (days 21, 28) phases. Salmonella enterica isolation and identification were completed by culture and PCR methods. The microbroth dilution method with SensititreTM (ThermoFisher Scientific, Lenexa, KS, USA) plates was used to determine the antimicrobial susceptibility and resistance of Salmonella strains. The susceptibility and resistance were interpreted based on the Clinical and Laboratory Standards Institute guidelines. The overall prevalence of Salmonella was 3.0% (43/1440). All isolates belonged to Salmonella enterica subsp. enterica serovar Typhimurium. Salmonella isolates were susceptible to azithromycin and resistant (100%) to ampicillin, streptomycin, sulfisoxazole, tiamulin, and tetracycline. Neither antibiotic, CTC or tiamulin, nor the route of administration, in-feed or in-water, had an effect (p > 0.05) on the occurrence of resistant Salmonella in the feces of piglets.

1. Introduction

Antimicrobial Resistance (AMR) is a global public health concern and is listed among the top health challenges in the world [1]. In the United States, 2.8 million AMR bacterial and fungal infections are estimated to occur, resulting in approximately 35,900 deaths every year [2]. The public health risks associated with antimicrobial use as growth promoters in food animal production have led to regulations to restrict antimicrobial use in many countries, including in the US. Antimicrobials have been used in swine routinely as a management tool by farmers [3]. The weaned (nursery) piglets consume most of these antimicrobials as they are more vulnerable to infectious diseases [4,5]. Several studies have shown a clear association between antimicrobial use and the development of AMR in food animals [6,7].
The use of In-feed antimicrobials for disease prevention has been an integral part of swine production and management. Tetracyclines, macrolides, and pleuromutilin are the most commonly used antimicrobials in swine [8]. Chlortetracycline (CTC), a tetracycline, is widely used in nursery pigs and is available for use by three routes of administration (in-feed, in-water, and injectable) [4,9,10]. Chlortetracycline (CTC) is used in nursery pigs to treat respiratory and enteric infections [11]. Tiamulin, a pleuromutilin and another commonly used antibiotic in nursery pigs to treat pneumonia and swine dysentery caused by Brachyspira hyodysenteriae, is administered in-feed or in-water [4,10]. Tiamulin is not considered medically important, does not require veterinary feed directives for its usage, and is likely to be frequently used or overused by swine producers.
Salmonella enterica is a foodborne pathogen of major public health concern. In the US, non-typhoidal Salmonella is estimated to cause annually about 1.35 million infections, 26,500 hospitalizations, and 420 deaths (https://wwwn.cdc.gov/norsdashboard/; accessed on 29 November 2023) and (https://www.cdc.gov/salmonella/index.html; accessed on 24 January 2024). Based on the CDC’s National Outbreak Reporting System (NORS), between 2008 and 2021, a total of 2904 Salmonella outbreaks have been reported in the US with 68,050 illnesses, 10,839 hospitalizations, and 109 deaths [12,13]. The US Department of Agriculture (USDA) Economic Research Service (ERS) reports the total annual cost of foodborne illnesses due to Salmonella is $3.6 billion [14]. Antimicrobial-resistant Salmonella is categorized as a serious threat level by the CDC and causes about 212,500 infections, resulting in about 70 deaths every year [2]. Considering its ubiquitous nature, Salmonella prevalence varies across the swine production systems [15]. Bernard-Roche et al. [16] reported a 36% prevalence of Salmonella among piglets.
Apart from implementing management systems to monitor and minimize antimicrobial use, optimizing the dosage and the route of administration can be an effective strategy to mitigate the development and dissemination of AMR without affecting the therapeutic efficacy of the antimicrobials. In swine, the oral route (in-feed and in-water) has been the most commonly used route of antimicrobial administration to treat pigs in groups [10,13]. Based on a systematic review, the oral administration of antimicrobials is considered to be a critical risk factor in the development and propagation of AMR in swine [17]. A study conducted in mice suggested that administering antimicrobials orally caused a notable increase in AMR in the gut microbiota compared to intravenous injection [18]. Another study in humans recommended that parenteral antibiotic administration is preferred over oral antibiotic administration as the gut commensal and pathogenic bacteria are less exposed to the antimicrobials, leading to less chance of resistance selection [19]. We hypothesize that a water-soluble antibiotic, administered in drinking water, is likely to be distributed more uniformly in the gut and that it, therefore, has a greater impact on the AMR of gut bacteria than the same antibiotic administered in a dry form mixed with the feed. Therefore, the route of administration of antimicrobials in food animal production is likely to be a significant contributing factor in the development of AMR. The impact of in-water vs. in-feed antibiotic administration on the development and persistence of AMR in the foodborne pathogen, S. enterica, has not been studied in nursery pigs. Therefore, the objective of this study was to assess the impact of the in-feed and in-water administration of CTC and tiamulin on the prevalence and AMR profiles of S. enterica in the feces of piglets.

2. Materials and Methods

2.1. Animals and Experimental Design

The experimental protocols used in this study were approved by the Kansas State University Institutional Animal Care and Use Committee (IACUC # 4033). The study was conducted at a commercial swine research facility in the Midwest. The facility is totally enclosed, environmentally controlled, and mechanically ventilated. Pens were equipped with slatted flooring and deep pits for manure storage. Each pen consisted of a six-hole stainless steel self-feeder and a pan waterer to provide ad libitum access to feed and water. A total of 1296 weaned piglets (L337 × 1050, PIC, Hendersonville, TN, USA) were used in the study. Piglets, weaned at 21 days of age, were allocated into 48 pens (27 piglets per pen) distributed in a single room so that average pen body weights were relatively equal among pens. After a 7-day acclimatization period, pens were randomly assigned to the following six treatment groups: (a) Control: piglets fed a basal diet with no antibiotic administration, (b) In-feed CTC: piglets fed the basal diet supplemented with CTC to provide 22 mg/kg body weight (BW; CTC-hydrochloride, Elanco-Eli Lilly and Company, Indianapolis, IN, USA) for 14 days, (c) In-water CTC: piglets fed the basal diet with CTC administered in drinking water to provide 22 mg/kg BW for 14 days, (d) In-feed tiamulin: piglets fed the basal diet supplemented with tiamulin to provide 5 mg/kg BW (Denagard®, Elanco Animal Health-Eli Lilly and Company, Greenfield, Indianapolis, IN, USA) for 14 days, (e) In-water tiamulin: piglets fed a basal diet with tiamulin administered in drinking water to provide 23 mg/kg body weight for 14 days, and (f) a combination of CTC and tiamulin (In-feed): supplemented with CTC to provide 22 mg/kg BW and tiamulin at 5 mg/kg BW for 14 days. Each treatment had 8 pens, and the treatment allocations to the pens followed a blocked design that ensured adjacent pens alternated among treatment groups. Also, the treatment allocation ensured that each pen treatment was in contact with an equal number of other pen treatments. The basal diet consisted of corn, soybean meal, vitamins, amino acids, and trace mineral supplements. Daily feed additions to each pen were accomplished through a robotic feeding system to provide measured amounts for individual pens. The duration of the study was 35 days, which was divided into three phases: pre-treatment (days 0 and 7), treatment (days 14 and 21), and post-treatment (days 28 and 35). In this study, all the antibiotic dosages and combinations are as per the FDA guidance for the treatment and prevention of diseases.

2.2. Fecal Sample Collection

Fecal samples were collected randomly from 5 piglets from each pen by a gentle rectal massage with proper restraining. Fecal samples were placed in individual plastic bags (Whirl-Pak® bags, Nasco, Ft. Atkinson, WI, USA) and transported on ice in a cooler to the Pre-harvest Food Safety Laboratory, Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, at Kansas State University and stored at 4 °C before processed within 24 h. The laboratory personnel were blinded to the treatments.

2.3. Isolation of Salmonella

Approximately, 10 g of fecal sample was added to 90 mL of Tetrathionate (TT) broth (Difco™, Sparks, MD, USA) with iodine added just before processing in high-temperature Nasco Whirl-Pak plastic bags (Nasco, Ft. Atkinson, WI, USA). The contents were mixed well and incubated at 37 °C for 24 h. One milliliter of TT broth was transferred to 9 mL of Rappaport Vassiliadis (RV) broth (Difco™) and incubated at 42 °C for 24 h. After enrichment, the RV broth was plated onto Hektoen Enteric (HE) agar (Difco™) and incubated at 37 °C for 24 h. A presumptive Salmonella colony (green colored with black center colonies) was picked and streaked onto a blood agar plate and incubated overnight at 37 °C. The Salmonella isolates from each sample were subjected to a rapid slide agglutination test using Salmonella antisera (BD Difco™ Salmonella O Antiserum Poly A-I & VI) for Salmonella identification. Fecal samples with presumptive Salmonella colonies and agglutination positive were considered Salmonella positive for further analyses.

2.4. PCR Identification of Salmonella enterica

DNA was extracted using the boil-prep method by suspending two bacterial colonies in 150 µL of nuclease-free water, followed by boiling for 10 min. The boiled bacterial lysate was centrifuged at 16,000× g for 2 min, and the resulting supernatant was used as a DNA template for PCR reactions. Species confirmation was completed by detecting invA (encodes for an inner membrane protein of the type III secretion system) and pagC (encodes for a periplasmic nonspecific phosphatase) genes using a duplex real-time PCR assay [20].

2.5. Antimicrobial Susceptibility Testing

Minimum inhibitory concentrations of antibiotics (MIC) to Salmonella isolates were determined using commercial Sensititre panel plates (SensititreTM CMV3AGNF, Trek Diagnostic Systems, Thermo Scientific Microbiology, Table 1). Bacterial colonies grown overnight on blood agar plates were suspended in demineralized water (Trek Diagnostic Systems), vortexed, and adjusted to 0.5 McFarland turbidity standard (Trek Diagnostic Systems, Thermo Scientific Microbiology). Ten microliters of the suspension were transferred to cation-adjusted Mueller-Hinton (Trek Diagnostic Systems) broth and vortexed. Then, 50 µL of the culture was inoculated into Sensititre plates and incubated at 37 °C for 24 h. The antimicrobials tested were amoxicillin/clavulanic acid, ampicillin, azithromycin, cefoxitin, ceftiofur, ceftriaxone, chloramphenicol, ciprofloxacin, gentamicin, nalidixic acid, streptomycin, sulfisoxazole, tiamulin, tetracycline, and trimethoprim/sulphamethoxazole. The Clinical and Laboratory Standards Institute (CLSI, 2023) guidelines for antimicrobial breakpoints were used to interpret the MIC values as either resistant (including intermediate) or susceptible. To obtain tiamulin MICs, the commercially prepared panels (SensititreTM BOPO7F, Trek Diagnostic Systems, Thermo Scientific Microbiology Table 1) were used. The veterinary standards were used to determine the resistance breakpoint for tiamulin since there are no values established by CLSI. Isolates that showed resistance to three or more antimicrobial classes were classified as multidrug-resistant (MDR) [21].

2.6. PCR Detection of Tetracycline Resistance Genes

The DNA extracted from Salmonella was screened for tet (A, B, C, D, and E) genes by PCR. E. coli positive control strains were procured from USDA (Meat Animal Research Center, Clay Center, NE, USA) and Dr. Marilyn C. Roberts at the University of Washington. The target genes and primers used are shown in Table 2. The 20 µL reaction volume consisted of 10 µL of the master mix, 6 µL of nuclease-free water, 1 µL each of forward and reverse primers, and 3 µL of the DNA template. Reactions were run in a Master-cycler gradient thermal cycler (Eppendorf, Hamburg, Germany), and the conditions were 1 cycle of initial activation at 95 °C for 3 min, 35 cycles of denaturation at 94 °C for 30 s, annealing at 60 °C for 90 s, and extension at 72 °C for 90 s, followed by a cycle of final extension at 72 °C for 10 min. Analysis of PCR products was completed by capillary gel electrophoresis in the QIAxcel Advanced system (Qiagen, Germantown, MD, USA).

2.7. Whole Genome Sequencing and Bioinformatics

DNA libraries from Salmonella isolates (n = 43) were prepared using the Illumina Nextera XT library preparation kit, with a modified protocol. The library quantity was assessed with Qubit (ThermoFisher). The libraries were then sequenced on an Illumina HiSeq platform 2 × 150 bp. Raw sequencing reads were trimmed and processed using BBDuk with a read quality trimming parameter of 22 for further downstream analysis [23]. The trimmed fastqs were assembled using SPAdes with the careful parameter [24]. The assembled contigs were then processed through the CosmosID core-genome SNP typing pipeline to evaluate the phylogenetic placement and SNP differences for meaningful epidemiological inferences. The CosmosID SNP typing pipeline uses Parsnp as the core-genome aligner to align the core genome of multiple microbial genomes [25]. While generating the core-genome alignment with the default parameters, Parsnp considers the recombination events and the variation that might exist within the microbial genomes. The final set of core-genome SNPs is then used by Parsnp to reconstruct the phylogenomic relationship among the genome using FastTree2 [26]. Bioinformatics tools from the Center for Genomic Epidemiology (CGE) were used to detect AMR genes (ResFinder; http://genepi.food.dtu.dk/resfinder, accessed on 20 January 2024) [27,28] and plasmids (PlasmidFinder; https://cge.food.dtu.dk/services/PlasmidFinder/, accessed on 20 January 2024) in the genomes [29].

2.8. GenBank Accession Numbers

Assembled genomes from 43 Salmonella isolates have been deposited in the National Center for Biotechnology Information (NCBI) under Bioproject Accession number PRJNA663574.

2.9. Statistical Analysis

Statistical analysis was performed using the Statistical Analysis Software (SAS version 9.4; Cary, NC, USA) LOGISTIC procedure with a sampling day in the STRATA statement and treatment group in the EXACT statement. Prevalence data were organized as a binomial response on the pen basis, i.e., the number of ‘+’ out of 5 samples per pen per sampling day. Because of the low prevalence rate, the exact conditional logistic regression approach was used to evaluate the treatment effect for Day 7 and Day 14 only. The block served as the stratifying variable for the conditional inference. Fixed effects of the model included treatment and the number of ‘+’ samples in the pen on Day 0. All tests were conducted at the 0.10 significance level. Comparisons between the treatment and control were carried out using the 2-sided exact test when the treatment group had ‘+’ samples and the 1-sided exact test when the treatment group had no ‘+’ samples [30]. The MIC data categorized as resistant or susceptible to 15 different antimicrobials were considered as binary responses and multidrug profiles. The MICs were expressed as median after the assumption of normality. A likelihood ratio chi-square test was performed to assess the relationship between the Salmonella prevalence and MDR profiles.

3. Results

3.1. Fecal Prevalence of Salmonella

A total of 1440 fecal samples were collected (240 fecal samples per week) during the study period. The overall prevalence of Salmonella was 3.0% (43/1440). The fecal prevalence of Salmonella within the treatment groups, treatment phases, and sampling week are displayed in Table 3. Neither antibiotic route of administration nor sampling week had an effect (p > 0.10) on the fecal prevalence of Salmonella. The prevalence of Salmonella was numerically higher among piglets in the in-water tiamulin (4.2%, n = 240) group compared to the control (2.5%), in-feed tiamulin (1.7%), in-feed CTC (2.1%), in-water CTC (3.8%), and in-feed combination of CTC and tiamulin (3.8%). The treatment phase had the numerically highest prevalence of Salmonella (5.0%, 24/480) when compared to the pre-treatment (3.5%, 17/480) and post-treatment phases (0.42%, 2/480), but the differences were not significant.

3.2. Prevalence of Tetracycline Resistance Genes

A tetracycline resistance gene PCR was conducted to see if chlortetracycline (CTC), one of the treatment groups in our experimental study, had any effect (positive or negative) on the prevalence of Salmonella. Salmonella isolates (n = 43) were screened for tetA, tetB, tetC, tetD, and tetE genes. The tetB gene was detected in all 43 isolates (100%). Only 11 (25.6%) and 4 (9.3%) isolates were positive for tetD and tetA genes, respectively, while all isolates were negative for tetC and tetE genes. Fifteen Salmonella isolates were positive for two tetracycline resistance genes (tetA and tetB), with 11 positives for tetB and tetD and four isolates positive for tetA and tetD. Only one isolate harbored three tetracycline-resistant genes (tetA, tetB, and tetD).

3.3. Antimicrobial Susceptibility Phenotypes

The phenotypic resistance and MIC distributions are summarized in a squashtogram as shown in Table 4. Neither CTC nor tiamulin treatment affected AMR profiles of Salmonella strains. All isolates were resistant (100%) to ampicillin, streptomycin, sulfisoxazole, tetracycline, and tiamulin, and the majority were resistant to ciprofloxacin (95.4%) and nalidixic acid (74.4%). All isolates were susceptible to azithromycin (100%) and cefoxitin (100%). A low level of resistance to trimethoprim/sulphamethoxazole (4.7%), ceftriaxone (7.0%), and ceftiofur (7.0%) was observed among the isolates. Resistance to most antimicrobials decreased (p < 0.10) over time. All isolates expressed resistance to at least three antimicrobial classes and, hence, were classified as MDR. Six unique resistance phenotypes were identified among the isolates. No isolate was resistant to all antimicrobials used in the susceptibility testing. The most common phenotype belonged to MDR strains, which were resistant to six antimicrobial classes (90.7%; 39/43), followed by resistance to seven antimicrobial classes (7.0%; 3/43), and only one isolate exhibited resistance to five antimicrobial classes (2%; 1/43). The most frequently observed (48.8%; n = 43) AMR phenotype was hepta-resistance pattern AMP_CIP_FIS_NAL_STR_TET_TIA (ampicillin, ciprofloxacin, streptomycin, sulfonamides, and tetracycline).

3.4. Whole Genome Sequence Results

The number of contigs ranged from 61 to 403, with the N50 ranging from 172,736–325,586 bp. The genome size ranged from 4,906,044 to 5,174,184 bp, with the GC content averaging 52.26%. The Parsnp in the CosmosID core-genome SNP typing pipeline identified all 43 isolates to be Salmonella enterica subsp. enterica serotype Typhimurium. ResFinder detected the presence of amikacin and tobramycin (aminoglycosides) (aac(6′)-Iaa), amoxicillin, ampicillin, cephalothin, piperacillin and ticarcillin (blaTEM-1B), ciprofloxacin (qnrB19), streptomycin (strA and strB), sulfamethoxazole and sulfisoxazole (sul2), and tetracycline (tetB) genes in all 43 isolates. None of the strains carried genes that encode for clavulanic acid, azithromycin, cefoxitin, ceftiofur, ceftriaxone, chloramphenicol, gentamicin, nalidixic acid, tiamulin, and trimethoprim. The correlation between the phenotypic and genotypic resistance to antimicrobials was over 90% for ampicillin, azithromycin, cefoxitin, ceftiofur, ceftriaxone, chloramphenicol, ciprofloxacin, streptomycin, sulfisoxazole, tiamulin, and tetracycline, as shown in Table 5. Furthermore, Plasmid Finder revealed the presence of Col (pHAD28) and IncQ1 in all strains as shown in Table 6. The low-level resistance to fluoroquinolone was attributed to no mutation in the plasmid-mediated quinolone resistance in the chromosome region. A low correlation was observed between amoxicillin/clavulanic acid (20.9%) and trimethoprim/sulfamethoxazole (4.7%). Despite having resistant genes to amoxicillin and sulfa, the isolates were rendered susceptible by the presence of clavulanic acid and trimethoprim in the two antimicrobials.

4. Discussion

In this study, we investigated the impact of in-feed and in-water CTC and/or tiamulin administrations on the fecal prevalence and AMR profiles of Salmonella enterica in weaned piglets. Salmonella is a major foodborne pathogen that colonizes the hindgut of swine and sheds in the feces and is a widely known cause of non-typhoidal infections in humans [31]. Antimicrobials are used to treat Salmonella infections in both humans and animals; therefore, an increase in AMR is of major concern. In the present study, we explored the possibility of using two methods of oral administration of antimicrobials in nursery piglets as a potential strategy to mitigate AMR. We tested our hypothesis using the two most commonly used antimicrobials in the swine industry, tetracyclines and tiamulin. Tetracyclines and tiamulin are administered mainly to treat respiratory diseases in nursery and grower pigs [10]. Chlortetracycline has more broad-spectrum activity, whereas tiamulin has a limited spectrum of activity against Gram-positive bacteria and mycoplasma [32]. At the global level, the swine industry utilizes the highest number of antimicrobials when compared to other food animals [33]. In the USA, the swine industry utilizes 42% by weight of all medically important antimicrobials approved for use in food-producing animals in 2019 [34]. Over 90% of these antimicrobials are administered orally in swine (both in-feed and in-water), making the oral route the most commonly used route of administration of antimicrobials in swine [31]. The oral administration of antimicrobials has a greater impact on the gut bacteria because of the direct exposure to antibiotics.
The effects of antimicrobials on gut bacteria are dependent on the dose administered, distribution of the antimicrobial, and concentration in the gut. Chlortetracycline forms insoluble complexes with aluminum, calcium, iron, and magnesium and also binds to lipids and proteins in the gut, thus reducing its absorption [35,36]. In an experimental study, the bioavailability of orally administered CTC in pigs was only 6% [37,38]. The low bioavailability indicates that most of the antibiotic remains in the gastrointestinal tract, at a concentration high enough to act on the microbes. In contrast, tiamulin bioavailability is up to 90% when administered orally (https://www.hpra.ie/img/uploaded/swedocuments/LicenseSPC_10825-017-001_06012016144953.pdf; accessed on 29 November 2023). Because oral bioavailability has an inverse relationship with intestinal concentration of the antibiotic [38], tiamulin was expected to be available at a minimal concentration (<10%) in the gut content. Interestingly, tiamulin is quickly absorbed into the blood, gets metabolized in the liver, and is excreted via bile into the gut to accumulate in the ileum and colon contents at concentrations sufficient to exert its effect on gut microbes (http://www.octagon-services.co.uk/articles/therapeutics.pdf; accessed on 29 November 2023). Similar to CTC, increasing the amount of tiamulin provided orally enhances bioavailability and its ultimate concentration in the gut contents [39]. As per the FDA’s regulations, one of the antibiotics (CTC) can be fed for only 14 days either in-water or in-feed. So, this study mimics what actual producers practice in a real-world setting here in the US.
Salmonella infections in pigs vary from subclinical to clinical enteritis, which are often self-limiting. Estimates on the fecal prevalence of Salmonella in pigs of different ages differ considerably by production and management types, ranging from 3.5 to 33% in the USA [40,41]. This variation is also observed across different stages of development of pigs, where piglets have a low level of Salmonella shedding when compared to sows [42,43]. In our study, the prevalence of Salmonella in nursery piglets was 3%. All isolates belonged to the S. Typhimurium serotype. This is the first time a single serotype of Salmonella has been reported in a prevalence study. The study was conducted in an enclosed commercial farm, which explains the possibility of a single serotype circulating within farm piglets. S. Typhimurium is the most widespread serotype with zoonotic importance in swine production systems according to the study [44,45]. We performed a PCR analysis of tetracycline-resistant determinants to assess whether the use of CTC, one of the treatment groups in our study design, had any effect on the prevalence of tetracycline-resistant Salmonella. Five genes that encode resistance to tetracycline, tet (A, B, C, D, and G), have been identified in Salmonella [46]. The tetB, which encodes for an efflux pump, was present in all Salmonella isolates recovered from this study. Similar findings were reported in previous studies, where tetB and tetC were the most dominant tetracycline resistance determinants in Salmonella [47]. The tetB gene has the widest distribution among Gram-negative bacteria [32]. Moreover, it was observed in this study that CTC, tiamulin, or a combination of both had no effect on AMR in Salmonella. The reason is all strains obtained in this study were resistant to tetracycline and tiamulin. Several other studies have recorded high resistance to tetracyclines [46,48,49]. In this study, both CTC and tiamulin did not influence the prevalence and AMR in Salmonella in piglets. Similarly, the study found that pigs treated with CTC showed constant high shedding of S. Typhimurium during the treatment and post-treatment periods [50]. They concluded that CTC helped the establishment of S. Typhimurium in the gastrointestinal tract of pigs, making it a persistent shedder throughout the post-treatment phase. Moreover, there was no significant difference in the resistance to CTC when Salmonella strains were compared between antibiotic-free swine farms and farms using antimicrobials [51,52]. The lack of difference in resistance observed in those studies could be due to the effect of carryover from earlier times when antibiotics were used. Information on the influence of tiamulin on AMR Salmonella is limited, and this study provides the first insight.
Salmonella tends to persist and spread resistance to other bacteria through plasmids, making the spread of resistance possible even without the selection pressure of antimicrobials, according to the study [53]. Resistant Salmonella could have been passed vertically from sows, or horizontally from farm personnel and the environment, and persisted throughout the length of the experiment. Moreover, all Salmonella strains recovered in the present study were MDR. One previous study observed persistence and increase in MDR S. Typhimurium in the food chain in the analysis of the prevalence data between 1996 and 2016 [54]. This increased resistance to multiple antimicrobials could potentially cause Salmonella to evolve into strains that are difficult to treat [55]. Pigs infected with S. Typhimurium are often asymptomatic or develop mild enteritis but can transmit the bacteria to humans through direct and indirect contact according to [56]. The high occurrence of MDR Salmonella recorded in this study warns of the possible risk of human exposure. Third-generation cephalosporins, fluoroquinolones, and macrolides are recommended for antimicrobial therapy against salmonellosis in humans [57]. However, a high level of resistance to most of these important classes of antimicrobials observed in this study poses concern. Most of the Salmonella strains exhibited no to low-level resistance to ceftiofur and ceftriaxone (third-generation cephalosporins) and azithromycin (a macrolide) in our study. Resistance to fluoroquinolones is low only due to an acquired resistance gene or genes and not attributed to mutations.
In recent years, WGS has emerged as an essential tool to identify AMR determinants. The tool has played a key role in providing information on epidemiological surveillance of AMR pathogens and studying the emergence of AMR in these pathogens, building new and improving present diagnostic tools, and developing novel antimicrobials of therapeutic importance [58]. Based on WGS, our results suggest that a similar clone of Salmonella enterica subsp. enterica serotype Typhimurium was circulating in the farm. The clone may have been dispersed among piglets and between pens through farm personnel and equipment. The persistence of the Salmonella enterica serotype Typhimurium DT12 in the pig farm environment for a long period of time that posed a risk of transmission to animals and humans was observed in [59]. S. Typhimurium has replaced S. Choleraesuis as the predominant serotype in pigs in the USA according to [60]. When genome sequences were compared with their expected AMR genotypes obtained by PCR, only tetB matched at 100%. Both tetA and tetD genes were not found in their genomes. The observed discrepancy might be due to the fact that PCR amplifies a small region in DNA that might be similar to the gene of interest but different from the actual entire sequence identified by WGS. Also, the plasmid carrying the resistant genes could have been lost through multiple re-streaking of the bacterial cells. High correlations between phenotypic and genotypic resistance to other antimicrobials were observed in this study. The discrepancy observed between some of the phenotypic and genotypic resistances could be attributed to a non-functional resistance gene, intrinsic resistance of the bacterial strains to some antimicrobials, and the inclusion of intermediate MIC values as resistant.
In summary, our results revealed that in-feed and in-water routes of antimicrobial administration did not influence the fecal prevalence and antimicrobial susceptibilities of Salmonella in this population of nursery piglets. Interestingly, the only serotype prevalent was S. Typhimurium. The lack of an AMR response was likely because of the widespread prevalence of AMR during the pretreatment phase. The MDR S. Typhimurium was resistant to several important and critically important antimicrobials to humans. Among the swine production stages, the nursery stage is a high-susceptibility period necessitating the implementation of an effective mitigation strategy to prevent the transmission of Salmonella infection. Our research findings do support the efforts of pork producers in enhancing the economic well-being of their herds and advancing the welfare of pigs across the swine production systems, thereby contributing to both public health and industry.

Author Contributions

Conceptualization, R.G.A. and T.G.N.; methodology, R.G.A., T.G.N. and M.D.T.; software, R.G.A. and Q.K.; formal analysis, R.G.A., V.L.I. and Q.K.; investigation, R.G.A. and V.L.I.; resources, R.G.A., T.G.N., M.D.T., J.D. and R.D.G.; writing—original draft preparation, V.L.I. and R.G.A.; writing—T.G.N., M.D.T., J.W., R.D.G. and J.D.; supervision, R.G.A.; project administration, R.G.A.; funding acquisition, R.G.A. and T.G.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Pork Board, grant number 19-028. The research was also supported in part by the USDA National Institute of Food and Agriculture, Hatch/Multistate Project 1014385.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Contribution no. 24-074-J Kansas Agricultural Experiment Station, Kansas State University, Manhattan, KS, USA.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. WHO. Urgent Health Challenges for the Next Decade. 2020. Available online: https://www.who.int/news-room/photo-story/photo-story-detail/urgent-health-challenges-for-the-next-decade (accessed on 23 January 2024).
  2. CDC. Antibiotic Resistance Threats in the United States; U.S. Department of Health and Human Services, CDC: Atlanta, GA, USA, 2019; p. 114. [Google Scholar]
  3. Aarestrup, F.M. The livestock reservoir for antimicrobial resistance: A personal view on changing patterns of risks, effects of interventions and the way forward. Phil. Trans. R. Soc. B 2015, 370, 20140085. [Google Scholar] [CrossRef]
  4. Davies, P.R.; Singer, R.S. Antimicrobial use in wean to market pigs in the United States assessed via voluntary sharing of proprietary data. Zoonoses Public Health 2020, 67, 6–21. [Google Scholar] [CrossRef] [PubMed]
  5. Lekagul, A.; Tangcharoensathien, V.; Yeung, S. Patterns of antibiotic use in global pig production: A systematic review. Vet. Anim. Sci. 2019, 7, 100058. [Google Scholar] [CrossRef] [PubMed]
  6. Bronzwaer, S.L.A.M.; Cars, O.; Buchholz, U.; Mölstad, S.; Goettsch, W.; Veldhuijzen, I.K.; Kool, J.L.; Sprenger, M.J.W. The Relationship between Antimicrobial Use and Antimicrobial Resistance in Europe. Emerg. Infect. Dis. 2002, 8, 278. [Google Scholar] [CrossRef] [PubMed]
  7. Tang, K.L.; Caffrey, N.P.; Nóbrega, D.B.; Cork, S.C.; Ronksley, P.E.; Barkema, H.W.; Polachek, A.J.; Ganshorn, H.; Sharma, N.; Kellner, J.D.; et al. Restricting the use of antibiotics in food-producing animals and its associations with antibiotic resistance in food-producing animals and human beings: A systematic review and meta-analysis. Lancet Planet. Health 2017, 1, e316–e327. [Google Scholar] [CrossRef] [PubMed]
  8. Holman, D.B.; Chénier, M.R. Impact of subtherapeutic administration of tylosin and chlortetracycline on antimicrobial resistance in farrow-to-finish swine. FEMS Microbiol. Ecol. 2013, 85, 1–13. [Google Scholar] [CrossRef] [PubMed]
  9. Apley, M.D.; Bush, E.J.; Morrison, R.B.; Singer, R.S.; Snelson, H. Use estimates of in-feed antimicrobials in swine production in the United States. Foodborne Pathog. Dis. 2012, 9, 272–279. [Google Scholar] [CrossRef] [PubMed]
  10. NAHMS Swine Study. Antimicrobial Use and Stewardship on U.S. Swine Operations. 2017. Available online: https://www.aphis.usda.gov/animal_health/nahms/downloads/amu-feedlots.pdf (accessed on 15 November 2023).
  11. Phillips, I.; Casewell, M.; Cox, T.; De Groot, B.; Friss, C.; Jones, R.; Nightingale, C.; Preston, R.; Waddell, J. Does the use of antibiotics in food animals pose a risk to human health? A critical review of published data. J. Antimicrob. Chemother. 2004, 53, 28–52. [Google Scholar] [CrossRef]
  12. CDC. Available online: https://wwwn.cdc.gov/norsdashboard/ (accessed on 19 November 2023).
  13. CDC. 2021. Available online: https://www.cdc.gov/salmonella/index.html (accessed on 24 January 2024).
  14. USDA ERS. Available online: https://www.ers.usda.gov/data-products/cost-estimates-of-foodborne-illnesses/ (accessed on 19 November 2023).
  15. Shawn, M.D.B. Salmonella in Swine: Prevalence, Multidrug Resistance, and Vaccination Strategies. Annu. Rev. Anim. Biosci. 2022, 10, 373–393. [Google Scholar] [CrossRef]
  16. Bernad-Roche, M.; Casanova-Higes, A.; Marín-Alcalá, C.M.; Cebollada-Solanas, A.; Mainar-Jaime, R.C. Salmonella Infection in Nursery Piglets and Its Role in the Spread of Salmonellosis to Further Production Periods. Pathogens 2021, 10, 123. [Google Scholar] [CrossRef]
  17. Sjolund-Karlsson, M.; Howie, R.L.; Blickenstaff, K.; Boerlin, P.; Ball, T.; Chalmers, G.; Duval, B.; Haro, J.; Rickert, R.; Zhao, S.; et al. Occurrence of β-Lactamase Genes Among Non-Typhi Salmonella enterica Isolated from Humans, Food Animals, and Retail Meats in the United States and Canada. Microb. Drug Resistance 2013, 19, 191–197. [Google Scholar] [CrossRef] [PubMed]
  18. Burow, E.; Simoneit, C.; Tenhagen, B.A.; Käsbohrer, A. Oral antimicrobials increase antimicrobial resistance in porcine E. coli—A systematic review. Prev. Vet. Med. 2014, 113, 364–375. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, L.; Huang, Y.; Zhou, Y.; Buckley, T.; Wang, H.H. Antibiotic administration routes significantly influence the levels of antibiotic resistance in gut microbiota. Antimicrob. Agents Chemother. 2013, 57, 3659–3666. [Google Scholar] [CrossRef]
  20. Rafii, F.; Sutherland, J.B.; Cerniglia, C.E. Effects of treatment with antimicrobial agents on the human colonic microflora. Ther. Clin. Risk Manag. 2008, 4, 1343–1357. [Google Scholar] [CrossRef]
  21. Bai, J.; Trinetta, V.; Shi, X.; Noll, L.W.; Magossi, G.; Zheng, W.; Porter, E.P.; Cernicchiaro, N.; Renter, D.G.; Nagaraja, T.G. A multiplex real-time PCR assay, based on invA and pagC genes, for the detection and quantification of Salmonella enterica from cattle lymph nodes. J. Microbiol. Methods 2018, 148, 110–116. [Google Scholar] [CrossRef]
  22. Schwarz, S.; Silley, P.; Simjee, S.; Woodford, N.; van Duijkeren, E.; Johnson, A.P.; Gaastra, W. Assessing the antimicrobial susceptibility of bacteria obtained from animals. Vet. Microbiol. 2010, 141, 1–4. [Google Scholar] [CrossRef]
  23. Miranda, C.D.; Kehrenberg, C.; Ulep, C.; Schwarz, S.; Roberts, M.C. Diversity of tetracycline resistance genes in bacteria from Chilean salmon farms. Antimicrob. Agents Chemother. 2003, 47, 883–888. [Google Scholar] [CrossRef]
  24. Bushnell, B. BBMap: A Fast, Accurate, Splice-Aware Aligner. In Proceedings of the 9th Annual Conference on Genomics of Energy & Environment Meeting, Walnut Creek, CA, USA, 17–20 March 2014; LBNL-7065E. Available online: https://escholarship.org/uc/item/1h3515gn (accessed on 21 January 2024).
  25. Bankevich, A.; Nurk, S.; Antipov, D.; Gurevich, A.A.; Dvorkin, M.; Kulikov, A.S.; Lesin, V.M.; Nikolenko, S.I.; Pham, S.; Prjibelski, A.D.; et al. SPAdes: A new genome assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. 2012, 19, 455–477. [Google Scholar] [CrossRef]
  26. Treangen, T.J.; Ondov, B.D.; Koren, S.; Phillippy, A.M. The Harvest suite for rapid core-genome alignment and visualization of thousands of intraspecific microbial genomes. Genome Biol. 2014, 15, 524. [Google Scholar] [CrossRef]
  27. Price, M.N.; Dehal, P.S.; Arkin, A.P. FastTree 2—Approximately Maximum-Likelihood Trees for Large Alignments. PLoS ONE 2010, 5, e9490. [Google Scholar] [CrossRef] [PubMed]
  28. Alessandra, C.; Ea, Z.; Aurora, G.F.; Mette, V.L.; Ole, L.; Laura, V.; Frank, M.A.; Henrik, H. In Silico Detection and Typing of Plasmids using PlasmidFinder and Plasmid Multilocus Sequence Typing. Antimicrob. Agents Chemother. 2014, 58, 7. [Google Scholar] [CrossRef]
  29. Zankari, E.; Hasman, H.; Cosentino, S.; Vestergaard, M.; Rasmussen, S.; Lund, O.; Aarestrup, F.M.; Larsen, M.V. Identification of acquired antimicrobial resistance genes. J. Antimicrob. Chemother. 2012, 67, 2640–2644. [Google Scholar] [CrossRef] [PubMed]
  30. Carattoli, A.; Hasman, H. PlasmidFinder and In Silico pMLST: Identification and Typing of Plasmid Replicons in Whole-Genome Sequencing (WGS). Methods Mol. Biol. 2020, 2075, 285–294. [Google Scholar] [CrossRef] [PubMed]
  31. Hirji, K.F.; Tan, S.J.; Elashoff, R.M. A quasi-exact test for comparing two binomial proportions. Stat. Med. 1991, 10, 1137–1153. [Google Scholar] [CrossRef] [PubMed]
  32. Crump, J.A.; Sjölund-Karlsson, M.; Gordon, M.A.; Parry, C.M. Epidemiology, clinical presentation, laboratory diagnosis, antimicrobial resistance, and antimicrobial management of invasive Salmonella infections. Clin. Microbiol. Rev. 2015, 28, 901–937. [Google Scholar] [CrossRef]
  33. Chopra, I.; Roberts, M. Tetracycline Antibiotics: Mode of Action, Applications, Molecular Biology, and Epidemiology of Bacterial Resistance. Microbiol. Mol. Biol. Rev. 2001, 65, 232–260. [Google Scholar] [CrossRef]
  34. Van Boeckel, T.P.; Brower, C.; Gilbert, M.; Grenfell, B.T.; Levin, S.A.; Robinson, T.P.; Teillant, A.; Laxminarayan, R. Global trends 501 in antimicrobial use in food animals. Proc. Natl. Acad. Sci. USA 2015, 112, 5649–5654. [Google Scholar] [CrossRef]
  35. FDA. 2019 Summary Report On Antimicrobials Sold or Distributed for Use in Food-Producing Animals. Center for Veterinary Medicine-FDA. 2019. Available online: https://www.fda.gov/media/144427/download (accessed on 15 November 2023).
  36. Neuvonen, P.J. Interactions with the absorption of tetracyclines. Drugs 1976, 11, 45–54. [Google Scholar] [CrossRef]
  37. Agwuh, K.N.; MacGowan, A. Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. J. Antimicrob. Chemother. 2006, 58, 256–265. [Google Scholar] [CrossRef]
  38. Nielsen, P.; Gyrd-Hansen, N. Bioavailability of enrofloxacin after oral administration to fed and fasted pigs. Pharmacol. Toxicol. 1997, 80, 246–250. [Google Scholar] [CrossRef]
  39. Peeters, L.E.J.; Daeseleire, E.; Devreese, M.; Rasschaert, G.; Smet, A.; Dewulf, J.; Heyndrickx, M.; Imberechts, H.; Haesebrouck, F.; Butaye, P.; et al. Residues of chlortetracycline, doxycycline and sulfadiazine-trimethoprim in intestinal content and feces of pigs due to cross-contamination of feed. BMC Vet. Res. 2016, 12, 1–9. [Google Scholar] [CrossRef]
  40. Riond, J.L.; Schreiber, F.; Wanner, M. Influence of tiamulin concentration in feed on its bioavailability inpiglets. Vet. Res. 1993, 24, 494–502. [Google Scholar]
  41. Davies, P.R.; Bovee, F.G.; Funk, J.A.; Morrow, W.E.; Jones, F.T.; Deen, J. Isolation of Salmonella serotypes from feces of pigs raised in a multiple-site production system. J. Am. Vet. Med. Assoc. 1998, 212, 1925–1929. [Google Scholar] [CrossRef]
  42. Rodríguez-Buenfil, J.C.; Alvarez-Fleites, M.; Segura-Correa, J.C. Incidence of salmonellosis and identification of serogroups and serotypes in a pig commercial farm in Yucatan. Rev. Latinoam. Microbiol. 2006, 48, 10–13. [Google Scholar]
  43. Dorr, P.M.; Tadesse, D.A.; Zewde, B.M.; Fry, P.; Thakur, S.; Gebreyes, W.A. Longitudinal study of Salmonella dispersion and the role of environmental contamination in commercial swine production systems. Appl. Environ. Microbiol. 2009, 75, 1478–1486. [Google Scholar] [CrossRef] [PubMed]
  44. Wilkins, W.; Rajić, A.; Waldner, C.; McFall, M.; Chow, E.; Muckle, A.; Rosengren, L. Distribution of Salmonella serovars in breeding, nursery, and grow-to-finish pigs, and risk factors for shedding in ten farrow-to-finish swine farms in Alberta and Saskatchewan. Can. J. Vet. Res. Rev. Can. Rech. Vet. 2010, 74, 81–90. [Google Scholar]
  45. Arguello, H.; Alvarez-Ordoñez, A.; Carvajal, A.; Rubio, P.; Prieto, M. Role of slaughtering in Salmonella spreading and control in pork production. J. Food Prot. 2013, 76, 899–911. [Google Scholar] [CrossRef] [PubMed]
  46. García-Feliz, C.; Ana, C.; Jesús Ángel, C.; Pedro, R. Herd-level risk factors for faecal shedding of Salmonella enterica in Spanish fattening pigs. Prev. Vet. Med. 2009, 91, 130–136. [Google Scholar] [CrossRef] [PubMed]
  47. Gargano, V.; Sciortino, S.; Gambino, D.; Costa, A.; Agozzino, V.; Reale, S.; Alduina, R.; Vicari, D. Antibiotic Susceptibility Profile and Tetracycline Resistance Genes Detection in Salmonella spp. Strains Isolated from Animals and Food. Antibiotics 2021, 10, 809. [Google Scholar] [CrossRef]
  48. Mendez, B.; Tachibana, C.; Levy, S.B. Heterogeneity of tetracycline resistance determinants. Plasmid 1980, 3, 99–108. [Google Scholar] [CrossRef] [PubMed]
  49. Pavelquesi, S.L.S.; de Oliveira Ferreira, A.C.A.; Rodrigues, A.R.M.; de Souza Silva, C.M.; Orsi, D.C.; da Silva, I.C.R. Presence of Tetracycline and Sulfonamide Resistance Genes in Salmonella spp.: Literature Review. Antibiotics 2021, 10, 1314. [Google Scholar] [CrossRef]
  50. Wang, X.; Biswas, S.; Paudyal, N.; Pan, H.; Li, X.; Fang, W.; Yue, M. Antibiotic Resistance in Salmonella Typhimurium Isolates Recovered From the Food Chain Through National Antimicrobial Resistance Monitoring System Between 1996 and 2016. Front. Microbiol. 2019, 10, 985. [Google Scholar] [CrossRef]
  51. Delsol, A.A.; Anjum, M.; Woodward, M.J.; Sunderland, J.; Roe, J.M. The effect of chlortetracycline treatment and its subsequent withdrawal on multi-resistant Salmonella enterica serovar Typhimurium DT104 and commensal Escherichia coli in the pig. J. Appl. Microbiol. 2003, 95, 1226–1234. [Google Scholar] [CrossRef] [PubMed]
  52. Ebner, P.D.; Mathew, A.G. Effects of antibiotic regimens on the fecal shedding patterns of pigs infected with Salmonella Typhimurium. J. Food Prot. 2000, 63, 709–714. [Google Scholar] [CrossRef]
  53. Gebreyes, W.A.; Thakur, S.; Morrow, W.E.M. Comparison of prevalence, antimicrobial resistance, and occurrence of multidrug-resistant Salmonella in antimicrobial-free and conventional pig production. J. Food Prot. 2006, 69, 743–748. [Google Scholar] [CrossRef]
  54. Bakkeren, E.; Huisman, J.S.; Fattinger, S.A.; Hausmann, A.; Furter, M.; Egli, A.; Slack, E.; Sellin, M.E.; Bonhoeffer, S.; Regoes, R.R.; et al. Salmonella persisters promote the spread of antibiotic resistance plasmids in the gut. Nature 2019, 573, 276–280. [Google Scholar] [CrossRef]
  55. Campioni, F.; Zoldan, M.M.; Falcão, J.P. Characterization of Salmonella Enteritidis strains isolated from poultry and farm environments in Brazil. Epidemiol. Infect. 2014, 142, 1403–1410. [Google Scholar] [CrossRef]
  56. Hoelzer, K.; Switt, A.I.M.; Wiedmann, M. Animal contact as a source of human non-typhoidal salmonellosis. Vet. Res. 2011, 42, 34. [Google Scholar] [CrossRef]
  57. Berbari, E.F.; Kanj, S.S.; Kowalski, T.J.; Darouiche, R.O.; Widmer, A.F.; Schmitt, S.K.; Hendershot, E.F.; Holtom, P.D.; Huddleston, P.M.; Petermann, G.W.; et al. 2015 Infectious Diseases Society of America (IDSA) Clinical Practice Guidelines for the Diagnosis and Treatment of Native Vertebral Osteomyelitis in Adults. Clin. Infect. Dis. 2015, 61, e26–e46. [Google Scholar] [CrossRef]
  58. Köser, C.U.; Ellington, M.J.; Peacock, S.J. Whole-genome sequencing to control antimicrobial resistance. Trends Genet. 2014, 30, 401–407. [Google Scholar] [CrossRef]
  59. Baloda, S.B.; Christensen, L.; Trajcevska, S. Persistence of a Salmonella enterica serovar typhimurium DT12 clone in a piggery and in agricultural soil amended with Salmonella-contaminated slurry. Appl. Environ. Microbiol. 2001, 67, 2859–2862. [Google Scholar] [CrossRef] [PubMed]
  60. Foley, S.L.; Lynne, A.M. Food animal-associated Salmonella challenges: Pathogenicity and antimicrobial resistance. J. Anim. Sci. 2008, 86, E173–E187. [Google Scholar] [CrossRef] [PubMed]
Table 1. The antimicrobial agents, concentrations, interpretive criteria, and WHO classifications applied for the study.
Table 1. The antimicrobial agents, concentrations, interpretive criteria, and WHO classifications applied for the study.
Antimicrobial AgentConcentration,
μg/mL
Breakpoint,
μg/mL 1
WHO
Classification 2
Amoxicillin/Clavulanic Acid1/0.5–32/16≥16/8Critically important
Ampicillin1–32≥16Critically important
Azithromycin 30.12–16N/ACritically important
Cefoxitin0.5–32≥16Highly important
Ceftiofur0.12–8≥4Critically important
Ceftriaxone0.25–64≥2Critically important
Chloramphenicol2–32≥16Highly important
Ciprofloxacin0.015–4≥0.12Critically important
Gentamicin0.25–16≥8Critically important
Nalidixic Acid0.5–32≥32Critically important
Streptomycin 42–64≥64Critically important
Sulfisoxazole16–256≥256Highly important
Tetracycline4–32≥8Highly important
Tiamulin 50.5–32≥32Important
Trimethoprim/Sulfamethoxazole0.12/2.38–4/76≥4/76Highly important
1 Resistance breakpoints based on Clinical and Laboratory Standards Institute (CLSI) guidelines for breakpoint (CLSI, 2023) except for streptomycin, azithromycin, and tiamulin, which have no CLSI breakpoints. 2 Categorization of antimicrobials for human medicine according to the World Health Organization (WHO, 2019). 3 CLSI has not established resistance breakpoints for azithromycin. 4 CLSI has not established resistance breakpoints for streptomycin; interpretive standards used are the National Antimicrobial Resistance Monitoring System (NARMS)-established breakpoints for resistance monitoring. 5 CLSI has not established resistance breakpoints for tiamulin; veterinary standards were used instead.
Table 2. PCR target genes and primers used for detection of tet (A, B, C, D, and E) resistance genes in Salmonella isolated from piglets administered with in-feed or in-water chlortetracycline and tiamulin.
Table 2. PCR target genes and primers used for detection of tet (A, B, C, D, and E) resistance genes in Salmonella isolated from piglets administered with in-feed or in-water chlortetracycline and tiamulin.
Target GenePrimer Sequences (5′ to 3′)Amplicon Size (bp)Source
tet(A)TET A1: CGA GCC ATT CGC GAG AGC2027[22]
TET A2: CGA AGC AAG CAG GAC CAT G
tet(B)TET BF: CAG TGC TGT TGT TGT CAT TAA576[22]
TET BR: GCT TGG AAT ACT GAG TGT AA
tet(C)TET C: TTG CAT GCA CCA TTC CTT GCG521[22]
TET CR: TGG TCG TCA TCT ACC TGC C
tet(D)TET D FW2: GGA TAT CTC ACC GCA TCT GC436[22]
TET D(RV1): CAT CCA TCC GGA AGT GAT AGC
tet(E)TET EF: TCC ATA CGC GAG ATG ATC TCC442[22]
TET ER: CGA TTA CAG CTG TCA GGT GGG
Table 3. Animal-level fecal prevalence of Salmonella enterica in piglets administered with in-feed or in-water chlortetracycline (CTC) and/or tiamulin (n = 1440).
Table 3. Animal-level fecal prevalence of Salmonella enterica in piglets administered with in-feed or in-water chlortetracycline (CTC) and/or tiamulin (n = 1440).
Treatment GroupTreatment Phases
Pre-TreatmentTreatmentPost-TreatmentTotal (%)
Week 1Week 2Week 3Week 4Week 5Week 6
Control1131006 (2.5)
In-feed CTC1220005 (2.1)
In-water CTC0341019 (3.8)
In-feed Tiamulin0310004 (1.7)
In-water Tiamulin03520010(4.2)
In-Feed CTC + Tiamulin0332019 (3.8)
Weekly Total (%)2 (0.8)15 (6.3)18 (7.5)6 (2.5)02 (0.8)
Total (%)17 (3.5)24 (5.0)2 (0.42)43 (3%)
Table 4. The MIC * distribution and percentage of resistance among Salmonella strains isolated from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin (n = 43).
Table 4. The MIC * distribution and percentage of resistance among Salmonella strains isolated from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin (n = 43).
AntimicrobialsResistant Breakpoint%
Resistant
0.0150.030.060.120.250.51248163264128256
Amoxicillin/Clavulanic Acid≥16/820.9 23227
Ampicillin≥16100 43
Azithromycin≥320 3139
Cefoxitin≥160 30112
Ceftiofur≥47 3134212
Ceftriaxone≥27 400000012
Chloramphenicol≥162.3 7351
Ciprofloxacin≥0.1295.4 11002120
Gentamicin≥820.9 2732227
Nalidixic Acid≥3274.4 12832
Streptomycin≥32100 43
Sulfisoxazole≥256100 43
Tetracycline≥8100 43
Tiamulin≥32100 43
Trimethoprim/sulphamethoxazole≥4/764.7 3431212
* MIC = Minimum inhibitory concentration. Note: The top dilution tested for each drug should be interpreted as ≥ and the lowest dilution tested for each drug as ≤. Phenotypes: AMP = ampicillin, CIP = ciprofloxacin, AUG = amoxicillin/clavulanic acid, AXO = ceftriaxone, CHL = chloramphenicol, FIS = sulfisoxazole, GEN = gentamicin, NAL = nalidixic acid, STR = streptomycin, SXT = trimethoprim/sulfamethoxazole, TET = tetracycline, TIA = tiamulin, XNL = ceftiofur.
Table 5. The association between the phenotypic resistance profiles and the resistance genes detected by whole genome sequencing of the Salmonella strains (n = 43) isolated from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin.
Table 5. The association between the phenotypic resistance profiles and the resistance genes detected by whole genome sequencing of the Salmonella strains (n = 43) isolated from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin.
Number of Resistant Strains
AntimicrobialPhenotype +/
Genotype +
Phenotype +/
Genotype −
Phenotype −/
Genotype +
Phenotype −/
Genotype −
Amoxicillin/Clavulanic Acid9 (20.9) 34 (79.1)
Ampicillin43 (100)
Azithromycin 43 (100)
Cefoxitin 43 (100)
Ceftiofur 3 (7) 40 (93)
Ceftriaxone 3 (7) 40 (93)
Chloramphenicol 1 (2.3) 42 (97.7)
Ciprofloxacin41 (95.3) 2 (4.7)
Gentamicin 9 (20.9) 34 (79.1)
Nalidixic Acid 32 (74.4) 11 (25.6)
Streptomycin43 (100)
Sulfisoxazole43 (100)
Tetracycline43 (100)
Tiamulin 43 (100)
Trimethoprim/sulphamethoxazole2 (4.7) 41 (95.3)
Table 6. A summary comparison of the phenotypic resistance profiles, PCR detection of the selected tetracycline genes, and the resistance genes detected by whole genome sequencing of Salmonella isolates (n = 43) recovered from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin.
Table 6. A summary comparison of the phenotypic resistance profiles, PCR detection of the selected tetracycline genes, and the resistance genes detected by whole genome sequencing of Salmonella isolates (n = 43) recovered from piglets administered with in-feed or in-water chlortetracycline and/or tiamulin.
Salmonella Strain* Phenotypic Resistance ProfilesPCR Detection of Resistance GenesResistance Genes Detection by WGS
120 A_2019-5-Salmonellaamp_axo_cip_fis_gen_nal_str_sxt_tet_tia_xnltetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
157 A_2019-5-Salmonellaamp_fis_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
265 A_2019-5-Salmonellaamp_cip_fis_gen_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
268 A_2019-5-Salmonellaamp_axo_cip_fis_gen_nal_str_sxt_tet_tia_xnltetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
286 A_2019-5-Salmonellaamp_cip_fis_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
288 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
309 A_2019-5-Salmonellaamp_aug_axo_cip_fis_gen_str_tet_tia_xnltetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
315 B_2019-5-Salmonellaamp_cip_fis_gen_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
317 A_2019-5-Salmonellaamp_cip_fis_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
325 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
329 A_2019-5-Salmonellaamp_cip_fis_gen_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
333 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
337 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetA, tetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
350 A_2019-5-Salmonellaamp_cip_fis_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
352 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
356 A_2019-5-Salmonellaamp_cip_fis_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
397 A_2019-5-Salmonellaamp_cip_fis_gen_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
502 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
503 B_2019-5-Salmonellaamp_chl_fis_gen_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
541 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
565 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
569 A_2019-5-Salmonellaamp_cip_fis_gen_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
602 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
612 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
624 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetA, tetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
633 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
637 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
642 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetA, tetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
643 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
647 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
648 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetB, tetDaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
669 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
685 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
693 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
720 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
815 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
867 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
884 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
888 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
900 A_2019-5-Salmonellaamp_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
931 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
1387 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
1399 A_2019-5-Salmonellaamp_aug_cip_fis_nal_str_tet_tiatetA, tetBaac(6′)-Iaa, strA, strB, blaTEM-1B, qnrB19,sul2, tetB
* Phenotypes: amp = ampicillin, cip = ciprofloxacin, aug = amoxicillin/clavulanic acid, axo = ceftriaxone, chl = chloramphenicol, fis = sulfisoxazole, gen = gentamicin, nal = nalidixic acid, str = streptomycin, sxt = trimethoprim/sulfamethoxazole, tet = tetracycline, tia = tiamulin, xnl = ceftiofur.
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

Ishengoma, V.L.; Amachawadi, R.G.; Tokach, M.D.; Kang, Q.; Goodband, R.D.; DeRouchey, J.; Woodworth, J.; Nagaraja, T.G. The Impact of In-Water vs. In-Feed Chlortetracycline and Tiamulin Administration in Piglets on the Fecal Prevalence and Antimicrobial Resistance of Salmonella. Appl. Microbiol. 2024, 4, 297-311. https://doi.org/10.3390/applmicrobiol4010020

AMA Style

Ishengoma VL, Amachawadi RG, Tokach MD, Kang Q, Goodband RD, DeRouchey J, Woodworth J, Nagaraja TG. The Impact of In-Water vs. In-Feed Chlortetracycline and Tiamulin Administration in Piglets on the Fecal Prevalence and Antimicrobial Resistance of Salmonella. Applied Microbiology. 2024; 4(1):297-311. https://doi.org/10.3390/applmicrobiol4010020

Chicago/Turabian Style

Ishengoma, Victor L., Raghavendra G. Amachawadi, Mike D. Tokach, Qing Kang, Robert D. Goodband, Joel DeRouchey, Jason Woodworth, and Tiruvoor G. Nagaraja. 2024. "The Impact of In-Water vs. In-Feed Chlortetracycline and Tiamulin Administration in Piglets on the Fecal Prevalence and Antimicrobial Resistance of Salmonella" Applied Microbiology 4, no. 1: 297-311. https://doi.org/10.3390/applmicrobiol4010020

Article Metrics

Back to TopTop