Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access September 15, 2022

Animal waste antibiotic residues and resistance genes: A review

  • Yi Wang , B. Dharmaveer Shetty , Sundaram Kuppu and Pramod K. Pandey EMAIL logo
From the journal Open Agriculture

Abstract

Antibiotic resistance is an emerging risk for human and animal health, and mitigating the risk requires an improved understanding of various sources of risks and identifying the level of threats for each source. Many antibiotics are currently used against pathogens for treating infections in animals and humans, and it is considered that antibiotic resistance genes (ARGs) acquired by pathogens may have an environmental origin. Because of contamination in ambient waterways, it is likely that ARGs may affect both organic and non-organic farming. While health risk as a consequence of ARGs is difficult to determine because of lack of understanding of dose-response, the presence of ARGs in human waste and animal manure, and the subsequent application of these organic wastes as fertilizers has a potential of spreading ARGs in the environment. Additional research is needed to understand the presence, growth, and transport of ARGs through animal wastes such as dairy manure. In this review, we focus on synthesizing information regarding the occurrence of ARGs in dairy manure, potential transport pathways, and factors responsible for the spread of ARGs in the environment. Besides, we also explore potential treatment methods that may contribute to the ARG removal in dairy manure and help alleviate ARG contamination.

1 Introduction

Antibiotic resistance, which represents the ability of an antibiotic resistant bacteria to protect themselves against antibiotic drugs, has become one of the major health concerns around the world due to an increasing number of infections and its related economic cost [1,2]. The risk caused by antibiotic resistance is considered to be one of the major threats to global health [3,4].

Since the first commercial antibiotic agents were introduced in 1935 (sulfonamides) and 1942 (penicillin), hundreds of subsequent antibiotic agents were discovered and made clinically available to the public [5,6]. Newer antibiotic agents continue to be discovered, though the rate of discovery has decreased [7,8]. Along with the slowed down discovery, resistance to antibiotics is also being observed with increasing frequency. Since the 1950s, infections resistant to multiple antibiotics, i.e., multidrug resistance, has increasingly become a human and animal health concern [9,10]. Bacterial species with multiple antimicrobial resistance clades have been discovered, such as E. coli, Salmonella typhi, Clostridium difficile, and Staphylococcus aureus [11]. In recent years, the increase in antimicrobial resistance has superseded the pace of research and development of novel antibiotics [11,12]. This phenomenon, unless addressed in a timely and targeted manner, could pose significant public health concerns due to decreasing options for effective antimicrobial therapy, and could reverse decades of advances since the advent of antibiotics.

In 2014, more than 700,000 human deaths occurred globally attributed to antibiotic resistant infections, and the cumulative economic damage in healthcare and reduced productivity would reach 100 trillion USD by 2050 [13]. Based on a report from the US. Centers for Disease Control and Prevention (CDC), it is estimated that more than 2.8 million antibiotic-resistant infections and 35,000 deaths occur annually due to antibiotic-resistance (CDC, 2019). At the present rate, it is predicted that by 2050 more than 10 million people may die annually due to antibiotic-resistant infections in the world [13].

The role of the environment is critical towards the pathogen’s ability to gain antibiotic resistance [14,15]. Antibiotic drugs are widely, and sometimes, indiscriminately used in animal farms for the treatment and prevention of bacterial diseases and occasionally, as a growth supplement [16]. Previous research has shown that extensive and indiscriminate use of antibiotics in animal husbandry can result in the creation of antibiotic resistant genes (ARGs) in livestock manure [17,18]. Treated livestock manure is used to fertilize cropland, which could aid in the subsequent spread of ARGs into the environment, leading to human and animal health challenges [19]. The goal of this article is to summarize and review previous studies on the presence, distribution, fate, and transport of ARGs in dairy manure, and the impacts of various manure treatment methods towards the reduction of such ARGs. Figure 1 shows the flow of ARGs from livestock waste and potential consequences on human health.

Figure 1 
               Livestock manure, manure application to cropland, and potential transfer of antibiotic resistances from manure to the ambient water and environment, and consequential public health risks.
Figure 1

Livestock manure, manure application to cropland, and potential transfer of antibiotic resistances from manure to the ambient water and environment, and consequential public health risks.

2 Classification of antibiotics and antibiotic resistance

2.1 Classification of antibiotics

Antibiotic agents can be classified into different groups based on their mechanism of action, and classification based on mechanisms’ clinical purposes are shown in Table 1. Depending on the mechanism of action, antibiotics affect bacteria in many ways, such as the inhibition of cell wall synthesis, cell membrane function, protein synthesis, nucleic acid synthesis, and/or other metabolic processes [20,21,22]. For example, β-lactam antibiotics target the penicillin-binding proteins (PBPs), which are responsible for the synthesis of the peptidoglycan layer of bacterial cell walls. Failure to form bacterial cell wall leads to bacterial death due to autolytic hydrolases.

Table 1

Different classes of antibiotics and clinical use for dairy animals

Mechanism Classes Spectrum of activity Effect on bacteria Clinical use for dairy animals Reference
Inhibition of cell wall synthesis β-lactam antibiotics Broad-spectrum/narrow-spectrum Bactericidal Mastitis, respiratory disease, diarrhea, uterine, metritis, foot rot, black leg, rhinitis, pneumonia, cellulitis, bursitis, and navel infection [59,60,65,206]
Glycopeptides Narrow-spectrum Bactericidal
Inhibition of cell membrane function Polymixins Narrow-spectrum Bactericidal Diarrhea and uveitis [59,60]
Lipopeptides Broad-Spectrum Bactericidal
Inhibition of protein synthesis (30 s Subunit) Aminoglycosides Narrow-spectrum Bactericidal Mastitis, diarrhea, locomotion, and bacterial enteritis [60,206]
Tetracyclines Broad-spectrum Bacteriostatic Respiratory disease, uterine, enteritis, weight gain, pneumonia, foot rot, metritis, and arthritis [59,60,206]
Inhibition of protein synthesis (50 s Subunit) Amphenicols Broad-spectrum Bacteriostatic Respiratory disease, pneumonia, and foot rot [60,65,206]
Macrolides Narrow-spectrum Bacteriostatic Mastitis and respiratory disease [60]
Lincosamides Narrow-spectrum Bacteriostatic Mastitis [206]
Streptogramins Narrow-spectrum Group A or Group B – Bacteriostatic; Group A & Group B – Bactericidal
Oxazolidinones Broad-spectrum Bacteriostatic
Inhibition of nucleic acid synthesis Quinolones Broad-spectrum Bactericidal Respiratory disease, diarrhea, and mastitis [59,60]
Aminocoumarin Narrow-spectrum Bacteriostatic/bactericidal Mastitis [206]
Ansamycins Broad-spectrum Bactericidal Mastitis (limited use) [207]
Inhibition of folic acid synthesis Sulfonamides Broad-spectrum Bacteriostatic Fever, bacterial pneumonia, calf diphtheria, diarrhea/digestive, and foot rot [65,206]
Diaminopyrimidines (trimethoprim) Broad-spectrum Bacteriostatic Diarrhea, coccidiosis, and systemic infections of newborn ruminants [59]

According to their spectrum of activity, antibiotics are classified as broad spectrum and narrow spectrum agents [23,24]. Broad spectrum antibiotics, such as tetracyclines, chloramphenicols, and cephalosporins (3rd, 4th, and 5th generation), can act against different classes of bacteria, including gram-positive and gram-negative bacteria. Narrow spectrum antibiotics such as penicillin, lincosamides, glycopeptides, streptogramins, and rifamycin [23] affect only a limited number of bacterial species. While antibiotics such as glycopeptides, lincosamides, macrolides, and streptogramins are used against gram-positive bacteria, other antibiotics such as aminoglycosides are only effective against gram-negative aerobic and facultative anaerobic bacilli [25]. In 2019, the United States Department of Agriculture in association with the World Organization for Animal Health, organized the Second International Symposium on “Alternative to Antibiotics.” Various studies have shown that vaccines can be a good alternative to antibiotics in food animals.

Based on the effect on bacteria, antibiotics are defined as bactericidal and bacteriostatic [23]. Antibiotics which prevent the growth of bacteria are considered bacteriostatic, whereas antibiotics which kill bacteria are known as bactericidal [26]. Bactericidal antibiotics such as β-lactams, aminoglycosides, glycopeptides, and quinolones cause bacterial cell death. Bacteriostatic drugs, such as chloramphenicol, oxazolidinones, sulfonamides, tetracyclines, and macrolides, inhibit bacterial growth. Some antibiotics possess both bactericidal and bacteriostatic abilities depending on the dose, duration of exposure, bacterial species, and other factors. For example, chloramphenicol triggers bactericidal activity against S. pneumoniae, and also initiates bacteriostatic activity against S. aureus [27,28].

2.2 Classification of antibiotic resistance mechanisms

There are four major types of bacterial resistance mechanisms. Bacteria may develop antibiotic resistance by reduction in permeability of cell membrane, enhancement of efflux pump efficiency, degradation of antibiotics, or/and modification of antibiotic targets. Antibiotic resistance mechanisms and corresponding bacteria genera is described in Table 2.

Table 2

Antibiotic resistance mechanisms and bacteria genera

Antibiotic classes Resistance type Bacteria genera Common mechanism Reference
Aminoglycoside Reduced permeability Gram-negative bacteria (e.g., Salmonella, Pseudomonas, Escherichia) and Mycobacterium Innate mechanism of resistance due to multi-layered cell walls, down regulation of porins [208]
Efflux pump Many gram-negative bacteria (e.g., Enterobacteriaceae, Salmonella, Pseudomonas, and Escherichia) Intrinsic AcrAD-TolC-type efflux pumps [209]
Degradation of antibiotics Staphylococcus, Pseudomonas, Serratia, and Mycobacterium AG-modifying enzymes [210]
Target modification Pseudomonas, Klebsiella, and Citrobacter 16S ribosomal RNA methyltransferases (RMTases) [211]
β-lactams Degradation of antibiotics Escherichia, Klebsiella, Enterobacteriaceae, Pseudomonas, Serratia, Enterobacter, and Staphylococcus β-lactamases (ESBLs, plasmid-mediated AmpC enzymes, and carbapenemases) [212,213,214]
Target modification Staphylococcus, Enterococcus, Streptococcus, and Neisseria Low-affinity PBP2a encoded by SCCmec, low-affinity PBP5 [215]
Chloramphenicol Reduced permeability Haemophilus, Burkholderia, and Salmonella Loss of an outer membrane protein [216]
Efflux pump Pseudomonas, Escherichia, Salmonella, Klebsiella, Staphylococcus, and Bacillus Specific exporters, E-1–E-8, multidrug transporter (MdfA, AcrAB-TolC) [216]
Target modification Escherichia and Bacillus Mutations in the major ribosomal protein gene [216]
Degradation of antibiotics Escherichia, Enterococcus, Staphylococcus, Bacillus, and Salmonella Chloramphenicol acetyltransferases [216]
Glycopeptide Target modification Enterococcus and Staphylococcus Reducing binding affinity by substituting terminal D-lactate or D-serine for D-alanine [215]
MLSB Efflux pump Corynebacterium, Enterobacter, Enterococcus, Gemella, Pseudomonas, and Streptococcus ATP-transporters or major facilitator transporters [217]
Degradation of antibiotics Pseudomonas, Serratia, Staphylococcus, Clostridium, Streptococcus, Enterobacter, Escherichia, and Klebsiella Inactivating enzymes including esterases, lyases, transferases, and phosphorylases [217]
Target modification Bacteroides, Enterococcus, Shigella, Staphylococcus, Salmonella, and Mycobacterium rRNA methylases [217]
Quinolone Reduced permeability Escherichia, Proteus, and Salmonella Altering expression of outer membrane porin proteins (OmpA, OmpF, and OmpC) [218,219]
Efflux pump Escherichia, Pseudomonas, Staphylococcus, Acinetobacter, and Stenotrophomonas Efflux systems (e.g., MdfA, NorA, MexXY, MexAB, MexCD, or MexEF) [219,220]
Target modification Escherichia, Klebsiella, Salmonella, Enterobacter, Citrobacter, Proteus, and Shigella Mutations in gyrA, gyrB, parC, and parE encoding the subunits of DNA gyrase and topoisomerase IV [219,220]
Sulfonamide Target modification Escherichia, Staphylococcus, Pneumocystis, Streptococcus, Campylobacter, Mycobacterium, and Neisseria Chromosomal mutations in the folP gene encoding DHPS, plasmid-borne resistance encoding DHPS variants [221]
Tetracycline Target modification Actinobacillus, Aerococcus, Streptococcus, Enterococcus, Psychrobacter, Staphylococcus, and Veillonella tet and otr genes code for ribosomal protection proteins (RPPs) [222]
Efflux pump Mycobacterium, Streptomyces, Acinetobacter, Clostridium, Escherichia, and Bacillus tet, otr, and tcr genes code for efflux pumps [222]
Degradation of antibiotics Escherichia and Bacteroides NADPH-requiring oxidoreductases [222]

2.3 Reduction in the permeability of antibiotics

One of the major strategies adapted by bacteria against antibiotics is to modify porin channels. While a cytoplasmic membrane in the gram-positive bacteria cell is surrounded by a thick layer of cell wall, a gram-negative bacteria cell consists of a thin cell wall, which is surrounded by a second lipid membrane [21]. This lipid membrane contains porins channels, which enables the passing of various molecules such as antibiotics [29,30]. The ability of bacteria to change porin channels allows them to protect themselves against antibiotics [31,32]. By modification of the expression/functionality of porin channels, bacteria may reduce the uptake of antibiotics. Many gram-negative bacteria resistant to β-lactams, fluoroquinolones, tetracycline, and chloramphenicol are known to modify protein channel to create resistance against antibiotics [33,34].

2.4 Expulsion of the antibiotics from the cell by efflux pumps

Efflux pumps are considered to be transport proteins. These transport proteins are found in the cytoplasmic membrane of both gram-positive and gram-negative bacteria [35]. The efflux pumps expel selective antibiotics and toxic substrates including lincosamides, macrolides, streptogramins, and tetracyclines from within cells into the external environment [36,37]. In some cases, mutations trigger amino acid changes which improve efflux pump efficiency in terms of extrusion of toxic substances such as antibiotics. For instance, Vettoretti et al. [38] described an amino acid mutation of MexXY efflux pump in P. aeruginosa, which results in an increased resistance to aminoglycosides, fluoroquinolones, and the β-lactam cefepime.

2.5 Modification and degradation of antibiotics

In addition to the use of efflux pumps, and modified porin channels, bacteria also develop other mechanisms such as the direct inactivation of antibiotics to protect themselves. Major classes of enzymes that inactivate antibiotics include β-lactamases, aminoglycoside-modifying enzymes, and chloramphenicol acetyltransferases [39]. β-lactamase is also known as penicillinase and was discovered in the 1940s [40]. Enzymes such as the β-lactamase break the β-lactam ring by hydrolysis, and inactivate the antibacterial properties of penicillin, cephalosporins, monobactams, and carbapenems [21,41]. The inactivated β-lactams are unable to bind to PBPs and thereby the process of cell wall synthesis is protected [41,42].

2.6 Modification of target sites in bacteria cells

The change in target sites of antimicrobials is another mechanism used by bacteria to prevent drug binding and resist against antibiotics [43]. Modification of critical target sites of antimicrobials generally leads to the inhibition of binding [21,44,45]. Studies have shown that changes in the ribosomes of Mycobacterium spp. resulted in a reduced affinity of streptomycin to target sites [46]. The presence of mutation in penicillin binding proteins in methicillin-resistant S. aureus and Listeria monocytogenes creates resistance to β-lactams [47,48]. Alterations in cell wall precursor components in staphylococcus and enterococci inhibit the binding of vancomycin and teicoplanin [49,50]. Mutations in DNA gyrase and topoisomerase IV in S. aureus and S. pneumoniae lead to reduced activity of fluoroquinolones [51,52,53], and mutations in RNA polymerases of Mycobacterium tuberculosis lead to resistance to rifampicin [54,55,56].

3 Antibiotics in food animal

Antibiotics have been globally used in food animals to prevent, restrict, and treat infectious diseases in both developed and developing countries [13,57]. A potential pathway of ARGs in animal waste is shown in Figure 2. Examples of commonly used antibiotics include trimethoprim/sulfonamides, tetracycline, β-lactams, and polymyxin B (Table 1) [58,59,60]. In 2010, global antimicrobial use in food animal production was 63,151 tons, which is estimated to increase to 105,596 tons by 2030 [61]. More than 6 million kilograms of medically important antibiotics and 5.3 million kilograms of not-medically important antibiotics in the U.S. were used in animals to prevent, control, or treat diseases in 2019 [62]. The majority (41%) of medically important antibiotics (i.e., important for treating human disease) used in animal agriculture in the US were administered to beef and dairy cattle [62], although the swine and poultry industries are estimated to be major consumers of antibiotics [61]. Cattle production was the third largest livestock industry in the world after the swine and poultry (approximately 80 million tons) in 2019 [63]. Currently, there are more than 60,000 dairy operations (≈17.5 million dairy herds) in the United States [64], and most operations use antibiotics for the treatment, control, and prevention of diseases. In 2013, about 87% of US dairy farms used antibiotics to treat cow mastitis, whereas 25 and 4% of farms used medically important antibiotics to prevent disease or promote growth for weaned heifers and pregnant heifers, respectively [65].

Figure 2 
               A potential pathway of ARGs recycling in dairy manure management system.
Figure 2

A potential pathway of ARGs recycling in dairy manure management system.

The administration of antibiotics in animals can be for various purposes: therapeutic, metaphylaxtic, prophylaxtic, or as growth promotors [59]. For therapeutic purposes, antibiotic drugs are administered to clinically ill animals. For example, around 16% of lactating dairy cows in the US receive antibiotics to treat mastitis every year [66,67], while 15% of beef calves in feedlots receive antibiotics therapy for clinical respiratory disease [68]. Metaphylaxis applications involve the pre-administration of antibiotics to the whole herd after the diagnosis of infection in one or more animals, in order to reduce the spread of the infectious disease [69,70]. Prophylaxis applications involve the administration of antibiotics to an individual animal or animal group as a preventive measure, prior to the appearance of clinical signs or infectious disease [71,72,73]. For example, prophylactic intramammary antibiotics are infused in the teat canal at the end of the lactation period to prevent and control future mastitis. Prophylactic antibiotics include penicillin, cephalosporins, and other beta-lactam drugs [66,67], amongst others. In addition, prophylactic antibiotics are used in 10% of healthy calves to reduce the risk of anticipated respiratory disease outbreaks [68]. In swine and cattle feeding, the prophylactic use of antibiotics occurs most often during the weaning period [71,72].

In addition to the use of antibiotics for reducing infections, antibiotics are also used as growth promotors [74,75]. In US, the use of medically important antimicrobials for growth promotion in food-producing animals has been eliminated by the US Food and Drug Administration (FDA) since 2017. However, 45 out of 155 countries are still using antimicrobials as a growth promoter according to the World Organization for Animal Health (OIE 2019). It is reported that low and subtherapeutic doses (1/10 to 1/100 of the curative dose) of antibiotics (such as penicillin, procaine, and tetracycline) are administered to animals via food or water to increase the rate of growth in animals [76]. Kirchhelle [57] highlights the long history of regulatory failures that has led to global antibiotic crisis.

It is important that remaining 45 countries follow suit on the ban on use of antibiotics as growth regulators. Several potential modes of action are proposed by the use of antibiotics as growth promotors [77]. The growth promotors are considered to be responsible for inhibition of nonspecific subclinical disease and gut microbiota, increasing availability of nutrition supply, reduction in harmful metabolites produced by intestinal bacteria, and improvement in absorption of dietary nutrients caused by thinning the intestinal wall [77,78,79,80]. Better understanding the mechanism of action might be helpful in developing effective alternatives [81].

At present, there is an increasing trend toward restricted use of antibiotics in food animals. In a meta-analysis conducted by Tang et al. [82] which included 81 animal studies, they found that reduction in the use of antibiotics lead to a decrease in the prevalence of antibiotic resistant bacteria by about 15% and multi drug resistant bacteria by 24–32%. In a similar meta-analysis study, Scott et al. [83] found that limiting antibiotic use in food animals reduced antibiotic resistant microbes, but the magnitude of the effectiveness were not quantified. In another interesting meta-analysis, Smith-Spangler et al. [84] found that the risk of isolating bacteria resistant to three or more antibiotics was higher in conventional chicken compared to organic chicken.

3.1 Excretion of antibiotic residues and presence of ARGs in dairy manure

From one health perspective, an approach to find optimal health for humans, animals, plant, and environment, it is important to find a solution for antibiotics crisis [85]. The major concern is that antibiotics critical for humans are used as mass medication for animals at very high dose. Though antibiotics could be effective at very low levels, large doses are normally administered to animals to facilitate drug delivery [86]. A major portion of this increased dose (up to 90%) is excreted in active form through feces [87]. Antibiotic residues such as the residues of tetracycline, chlortetracycline, and oxytetracycline are the most commonly detected antibiotics in dairy manure [88]. A study by Wallace and Aga [88] detected tetracycline residues in dairy feces and found that the majority of residues were primarily attached to fecal solids. In contrast, the widely used β-lactams are relatively more soluble and appear to be less persistent in manure [18]. To illustrate, a study that collected more than 80 fresh manure samples from 11 dairy farms in the northeastern United States, which commonly used penicillin and cephalosporins in their animals, showed no detection of β-lactam residues in any of their manure samples [18]. At low concentrations, sulfonamide, macrolide, and lincosamide residues have also been detected in dairy manure samples [18]. The presence of antibiotic residue in manure has been shown to negatively impact several downstream processes.

Prevalence of ARGs in animal feces are associated with the extensive and indiscriminate use of antibiotics in food animals [17,89]. Under antibiotic selection pressure, antibiotic resistant bacteria can be selected in the animal gut. Antibiotic resistance can develop by gene mutations (vertical gene transfer) or acquire exotic genes (horizontal gene transfer) in the animal gut from other species, strains, or extracellular DNA [90]. Feeding milk with residual concentrations of antibiotics has been shown to result in increased antibiotic resistance in E. coli isolates in the fecal samples of pre-weaned calves [91]. Since dairy manure is considered to be a reservoir for more than 60 types of ARGs [92], ARGs are frequently detected in livestock waste including manure, wastewater, lagoon slurries, and sediments [93,94,95]. For example, fecal samples collected from three large commercial pig farms in China yielded tens of thousands of times more ARGs than those not exposed to antibiotics [96].

ARGs detected in animal manure include various types of genes: bla for the β-lactams [97,98]; van for glycopeptides [97,99], erm gene for macrolide-lincosamide-streptogramin B (MLSB) [97,98,100], sul genes for sulfonamides [97,98,100], tet genes for tetracyclines [97,98,100], satA/satG for streptogramin [101], fexA, cfr, cmlA, and floR for chloramphenicols [102], acc for aminoglycosides [97,98], fca for FCA (quinolone, florfenicol, chloramphenicol, and amphenicol) [97,100], mcr for colistin [94], van for vancomycin [97], and mdr for multidrug [92,97]. The most commonly detected ARG categories in animal waste included tet, sul, erm, fca, and bla, which correspond to the most commonly used antibiotic types in animal agriculture [95]. Among these five ARG categories, tet and sul genes were generally most abundant, present in almost all livestock wastes [95]. The considerable variation in ARGs may result from different usage of antibiotics across livestock types and farm locations [95].

3.1.1 Transfer of AR and ARGs into the environment

The presence of antibiotics and ARGs in animal manure is a concern, because the majority of manure produced in animal-agriculture system is used as fertilizers in croplands. The elevated levels of antibiotics in manure, and its subsequent use in cropland could potentially contaminate the environment. Horizontal gene transfer is mainly responsible for the spread of ARGs in the environment, including transformation, transduction, and conjugation [103]. ARGs are classified as two forms: intracellular DNA transmitted by conjugation and/or transduction [104,105] and extracellular DNA transferred through transformation [106]. Transformation involves the direct passage of free DNA conferring antibiotic resistance from nearby dead cells [107]. Transduction involves the transfer of DNA from one bacteria cell to another via phagocytosis [108]. Bacteriophages containing ARGs infect the cell and introduce genes into the receiving bacteria [108,109]. Conjugation is the major way of transport of ARGs [110,111,112]. About 80.8% of all tested strains are considered to transmit ARGs by conjugation [112]. This mechanism is mediated by plasmids and involves the transfer of circular DNA by sexual pilus which requires cell-to-cell contact.

ARGs may be associated with bacteriophages and prophages, plasmids, and transposable factors [113], which contribute to the spread of ARGs. A large number of mobile genetic elements (MGEs) such as integrons, transposons, and plasmids are detected in animal manure [96,114,115]. The abundance of ARGs in swine farms in China were found to be significantly correlated with intI1 (class 1 integrase gene) and IS6100-type transposons in manure samples [114].

The overuse of antibiotics in animal-agriculture system and the subsequent application of animal wastes into soil systems (e.g., through land application of manure) introduces antibiotic residues, antibiotic-resistant bacteria, and ARGs into this environment [116]. The soil environment is considered one of the largest reservoirs of microbial communities, and it plays a crucial role in the emergence and spread of ARGs (Figure 1). Agricultural soil is considered to be a hotspot for bacteria to exchange genetic material by horizontal gene transfer [117]. Studies have shown that extracellular ARGs are able to persist in soil and sediments for long periods of time, up to years [118]. Subinhibitory concentrations of antibiotics in soil could cause selection pressure, and bacteria expedite their genetic material exchange rates to enhance horizontal transfer of ARGs [119,120,121]. Besides antibiotic residues, environmental conditions can also trigger horizontal gene transfer via stress response mechanisms [120,122]. For example, heavy metals in animal feed as alternative biocides or animal growth promotors [123] can cause stress on bacteria and select for ARGs through co-resistance or cross-resistance [124].

These manure-associated ARGs can be transferred from cropping and grazing lands [125] to surface/ground waterways (Chee-Sanford et al. 2009) by rainfall and runoff. ARGs from the soil can enter the food chain via contaminated crops and groundwater and cycle back to humans [126,127]. A study found elevated levels of tetracycline resistance genes in groundwater underlying two swine operations [128]. Besides, ARGs from water and soil may also enter the air by physical movements [129]. Antibiotic resistant bacteria on airborne particles can be inhaled by humans through dust particles or can settle on skin surfaces [130,131,132]. ARGs in surface and ground water, soil, and air can proliferate by horizontal gene transfer to indigenous bacteria including pathogens, and eventually reach humans causing public health concerns [133,134,135]. One study stated that resistance genes can persist for over 10 years in the absence of the corresponding antibiotic agents [136]. Hill et al. [137] analyzed crop nutrients and environmental contaminants including antibiotics from nine uncomposted manure samples from Idaho dairies collected for over 2.5 years. Monensin was the most frequently detected antibiotics. They also investigated the relative abundance of several plasmids involved in the spread of ARGs and found that IncI, IncP, and IncQ1 in almost all the samples. Ruuskanen et al. [138] studied the effect of animal manure on dissemination of ARG to the farm environment in Finland. Their findings show that there was increased relative abundance of two ARGs (Sul1 and Tet m) in cow and swine manure that was stored in the farm for approximately 9 months. They hypothesized that the selection pressure could have been the reason for such an increase. The atmospheric environment of composting has been shown to harbor ARGs. Gao et al. [139] in their study found 22 ARGs and int1 in the air around composting. This necessitates the proper regulations for such a facility as the presence of ARGs in the air could pose risk of transfer to humans and animals in the vicinity of the facility breathing such contaminated air.

3.1.2 Transfer of AR and ARGs to crop plants

The time-tested healthy practice of using dairy waste as manure for crop plant has been challenged with the advent and use of antibiotics over the past 75 years. Animal manure is a rich source of essential organic nutrients for plant growth. The strategy of fertilizing plants with animal manure had a two-pronged advantage of using them as a cheap source of fertilizer and an economic way of disposing animal waste. Unfortunately, with the increasing use of antibiotics over the past 75 years and the emergence of antibiotic resistant microbes carrying ARGs in animal waste, regulatory agencies around the world have imposed restrictions on their use as fertilizer to several crops. The age-old healthy practice with two-pronged advantage has become a two-sided disadvantage. This has left us with a problem of finding alternatives for both disposing animal waste and reliance on inorganic fertilizer as a sole source of nutrients for many crops.

In a study to understand the effect of transfer of ARG to plants fertilized with manure-based amendments, Guron et al. [140] used controlled, integrated, and replicated greenhouse study. They used radish and lettuce, a leafy vegetable as their model plants. Radish, a root vegetable was found to carry a great load of ARG, and species richness compared to lettuce. Their finding on association shows that there is almost exclusive association between ARGs and MGEs in plants that receive raw manure amendments. Composting did alleviate the mobility of manure resistance derived traits. In another study, Zhang et al. [141] found that the absolute abundance of ARGs and MEGs on the rhizosphere and phyllosphere of soil fertilized with raw manure from poultry and cattle farms was significantly higher than the ones detected in root and shoot endophytes. Compared to the control samples, the number of ARGs and MEGs detected in shoot and root endophytes were significantly higher than the controls. This clearly shows that application of raw manure with ARGs and MEGs might affect the microbiome of vegetable crops.

In another study, Zhang et al. [142] found that manure application did not enrich ARGs in the root endophytes in cherry radish. Cattle manure application increased the ARG resistome of rhizosphere and phyllosphere microbiota but not the endophytic bacterial microbiota. Despite being buried in the soil, the endophytic microbiota was not significantly affected. This clearly shows that transfer of ARGs is crop dependent. More studies on transfer of ARGs from manure to different crops are required to precisely regulate the use of manure on different crops.

Zea mays (corn) is an economically important crop in the United States of America and around the world. Mullen et al. [143] found that antibiotic residues of sulfonamide and tetracycline bioaccumulated in plant shoots, when the plants were grown with dairy manure amended soil. When antibiotics spiked manure (1 mg/kg) was used, tetracycline and sulfamerazine were detected at concentrations of about 300 and 1,260 ng/g, respectively. Interestingly, no antimicrobial resistance genes were detected. Though, in this study, the presence of antibiotic residues in the shoot were studied, the accumulation of these residues in the corn cobs were not analyzed.

Zhu et al. [144], in their study on the phyllosphere of staple crops under pig manure fertilization, found that ARGs were prevalent in both of their crops of interest, rice and wheat. They were able to detect 162 unique ARGs and five MGEs from rice and wheat samples. They showed that cleaned (treated) pig manure led to lower enrichment of ARGs compared to raw manure. They also found that wheat samples had higher ARG diversity than rice.

Zhao et al. [145] studied the effect of long-term manure application on antibiotic uptake by crops. Peanuts on the field with long term (>15 years) manure application was studied. Antibiotics were found in all peanut tissues (root, stem, leaves, cells, and kernels). Consumption of those peanuts were considered to present moderate health risk to humans.

Due to the concern of pathogens, most farmers, growing leafy greens worldwide, currently refrain from using animal manure as fertilizer. Currently, the United States FDA has a regulation of 120 days interval between the application of raw manure and planting for the crops where the economically important part of the plant comes in contact with the soil and 90 days interval for the ones where it does not come in direct contact with the soil [146].

3.2 Treatment technologies to reduce ARGs

Manure is usually applied to agricultural land nearby due to its agronomic value. The application of manure as fertilizers may increase the spread of ARGs into the environment and eventually affect human health. Although existing manure management technologies used for treating manure are not designed to mitigate ARGs. Fate of ARGs during different treatments of manure are shown in Table 3.

Table 3

Fate of antibiotic resistant genes during different treatments of manure

Genes Changes in gene abundance Type of treatment Scale Feedstock Reference
ermB, ermF, and tetW Increased 0.2–1.5 log copies/g Mesophilic anaerobic digestion Laboratory-scale Wastewater sludge [180]
sulI, sulII, tetC, tetG, tetX, and intI1 Decreased 0.5–1.5 log copies/g
sulI, sulII, ermB, ermF, tetO, tetW, tetG, and intI1 Decreased 0.5–1.5 log copies/g Thermophilic anaerobic digestion Laboratory-scale Wastewater sludge [180]
aphA2 Decreased 2.2 log copies/g in summer and 1 log copies/g in winter Ambient temperature anaerobic digestion (summer and winter) Pilot-scale Dairy manure [190]
ermB Decreased 1 log copies/g in both seasons
bla TEM-1 Decreased 2.2 log copies/g in summer and 0.84 log copies/g in winter
tet (X, Q, W, G, C), erm (X, Q, F, B), and sul (1, 2) Decreased 1.03–4.23 log in total (copies/16S rRNA) Mesophilic anaerobic digestion Laboratory-scale Swine manure with wheat straw [175]
tet and erm Decreased 0.1–0.15 copies/16S rRNA Anaerobic digestion Field-scale Swine manure [177]
sul, fca, and aac 1.4–52 times increase (copies/16S rRNA)
sul2, intI1 and intI2, and gryA Decreased 0.1–1.5 log copies/g Moderate anaerobic digestion Laboratory-scale Dairy manure [181]
tetC, tetM, tetQ, tetX, and sul1 Increased 0.1–3.5 log copies/g
sul1, sul2, intI1 and intI2, gryA Decreased 0.1–1.5 log copies/g Mesophilic anaerobic digestion
tetC, tetM, tetQ, and tetX Increased 0.1–1 log copies/g
sul1, sul2, intI1 and intI2, gryA, tetM, tetX, tetW Decreased 0.1–1.5 log copies/g Thermophilic anaerobic digestion
tetC Increased 1 log copies/g
tetG Decreased <1 log copies/g Stockpiled Field-scale Dairy manure [223]
RPP tet No change
ermB, ermC, ermF, ermT, and ermX Decreased 1–2 log copies/g
tetA, tetC, and RPP tet Decreased ∼4 log copies/g Composted Field-scale Swine manure [224]
tetG No change
tet, sul, aac, erm, bla, mdr, fca, and van Reduced 5 times in total (copies/16S rRNA) Composted Field-scale Dairy manure [97]
erm genes (A, B, C, F, T, and X) Decreased ∼2–3 log copies/g Composted (55°C with aeration) Laboratory-scale Swine manure [156]
tet genes (A/C, G, M, O, T, and W) Decreased 1–4 log copies/g
erm genes (A, B, C, F, T, and X) Decreased < 1 log copies/g even increased slightly (ermF) Lagoon (room temperature) Laboratory-scale Swine manure [156]
tet genes (A/C, G, M, O, T, and W) Decreased < 1 log copies/g even increased slightly (tet(A/C))

3.2.1 Solid–liquid separation (SLS)

SLS is applied in many US dairy farms to remove fibrous solids from manure slurries to prevent accumulation in long time storages [18]. The separated solids can be used as a renewable bedding source, or as a feedstock for digestion processes [65,147]. Antibiotic residues have different solubility and sorption properties, and thus there is distinct partitioning between solid and liquid portions after SLS [88,148]. However, there are only a few articles studying the effect of SLS on the attenuation of ARGs [18], and the results showed a higher abundance of ARGs in separated solids compared with raw manure. Wallace et al. [149] collected raw and separated dairy manure from an SLS system located on a dairy farm with 1800 cows, and SLS treatment resulted in increased quantities of sul1, sul2, and tetO in separated solids. This suggests that mechanical separation without subsequent processing is likely to concentrate on certain types of ARGs in separated solids. Tien et al. [98] collected soil samples in field plots amended with untreated and mechanically dewatered manure, and authors reported that the occurrences of ARGs were more frequent in soil receiving raw manure than that receiving dewatered manure. Gros et al. [150] investigated the fate of ARGs in a full scale on farm livestock waste treatment plant. The plant used SLS, anaerobic digestion, and reverse osmosis of liquid digestate. Their findings show that antibiotics mostly partitioned onto the solid fraction after SLA. Zhang et al. [151] explored the change in ARGs in liquid and solid fractions of dairy manure. Results show that high total solids enhanced ARG reduction in liquid fraction, while those in solid fraction increased. Post treatment of such solid waste where ARGs increase should be a mandatory step to prevent their spread into the environment. Figure 3 elaborates flow of ARGs in dairy manure, and potential treatment options to control ARGs dissemination.

Figure 3 
                     Dairy manure management system and dairy waste management technologies for controlling the dissemination of ARGs into environment.
Figure 3

Dairy manure management system and dairy waste management technologies for controlling the dissemination of ARGs into environment.

In another study, Wright and Gooch [152] measured the partitioning of ARGs in an on-farm screw press separation. They found that the majority of antibiotic residue were partitioned with liquid stream. Ray studied the impact of on-farm SLS followed by rotary drum composting on the fate of ARGs. The authors studied sul1, bla OXA-1 , int1, and tetO. They found that about 88% of tetO were partitioned into separate solids, while 70% or more of sul1, bla OXA-1 , and int1 were partitioned into liquid. Rotary digestion process was effective in reducing tet0, sul1, bla OXA-1 , and int1 at 98, 75, 87, and 60%, respectively.

3.2.2 Composting

Composting is used to convert organic material into a stable, humus-rich material, which could generate heat and deactivate pathogens in the process. The effect of composting on reducing ARGs varies among different studies [153,154,155,156,157]. It was reported that reduction in ARG in intensively managed composts is generally more effective than stockpiling [158,159]. A previous study showed that composted manure had the lowest abundance of ARGs compared to untreated, anaerobically digested, and mechanically dewatered dairy manure from farms [98]. Xie et al. [160] suggested a significant reduction in the total abundance of ARG in cattle and poultry manures after thermophilic composting in a commercial scale. ARGs that confer resistance to vancomycin, sulphonamides, beta-lactams, and MLSB were reduced by several folds to more than one order of magnitude in cattle and poultry manure [160]. However, genes such as tetL, aadA, and aadA2 were persistent in composted material. It was reported that thermophilic composting removes ARGs better than mesophilic composting [95,161]. Additives such as clay, biochar, superabsorbent polymers, zeolite, calcium, and superphosphate can promote the effects of composting on ARGs [102,162,163,164]. Cui et al. [102] stated that biochar could change the fate of ARGs during the composting process by reducing the bioavailability and mobility of heavy metals, which could provide a suitable habitat for microbial proliferation by increasing the temperature and shifting the bacterial community.

Liao et al. [165] in their study showed that hyperthermophilic composting removed ARGs and MEGs more efficiently than conventional composting at 89% and 49%, respectively. The reduction in ARGs and MEGs were associated with the reduction in bacterial abundance. Their modeling using partial-least square path shows that reduction in MEGs also play a key role in the reduction in ARGs. Cao et al. [166] studied the profiles of ARGs at different stages of composting. In their study, they found that ARGs increased during composting. The same group, in another study, found that inoculation of composting material with microbial agents was shown to promote the removal of ARGs in pig manure [167]. Aerobic combined anaerobic composting has been shown to significantly reduce the abundance of ARGs in swine manure [168]. Yu et al. [169] have shown that integration of bioelectrochemical system into conventional anaerobic composting could enhance the reduction in ARGs. The attenuation of ARGs was attributed to the combination of CaO2 and bioelectrogenesis. Co-composting of swine manure with cauliflower and corn straw as a bulking agent was shown to significantly decrease the absolute abundance of most ARGs [170]. In an industrial scale composting, Tang et al. [171] found that the abundance of ARGs and MGES increased during aerobic thermophilic stage. Firmicutes and Actinobacteria were identified as potential hosts. MGEs in the host were identified as a hindering factor in ARG removal. Zhang et al. [172] showed that aerobic composting of cow manure was effective in reducing ARGs. In their study they identified Acinetobacter and Pseudomonas as potential hosts. After 14 days of aerobic composting, the abundance of ARGs were reduced by over 83%.

3.2.3 Anaerobic digestion

Anaerobic digestion is a process of decomposition, where organic compounds are degraded by microbial community in the absence of oxygen. Recent studies have demonstrated that anaerobic digestion is a potential way to remove ARGs [173,174,175]. However, other studies have indicated that the reduction effect of anaerobic digestion could be gene specific [95,100,176,177]. Some ARG classes were even enriched by anaerobic digestion. The inconsistent effect of anaerobic digestion on ARG removal can be attributed to complicated interactions among ARGs, bacteria, digestion parameters, and associated metabolic pathways in the anaerobic digestion process, which requires additional research [178]. Wallace et al. [149] investigated the abundance of sulfonamide- and tetracycline-resistant ARGs (log(ARG Copies/16S rRNA)) through a full-scale advanced anaerobic digester, and the authors found that only sul1 and sul2 copies were significantly reduced (P < 0.001) by 5 and 10%, respectively. However, tetracyclines-related genes (tetW and tetO) were unchanged in this study.

Previous studies have investigated the reduction in ARGs during anaerobic digestion under various conditions such as temperature [174,179,180,181], retention time [180], pH [182], co-digestion [175,183], antibiotics [184,185,186], and pretreatments [180]. Temperature could shift the diversity, function, and interactions of the microbial community [187]. Bacterial communities can be one of the major drivers in ARG reduction during anaerobic digestion [175,181]. Some bacteria in livestock manure are potential hosts of ARGs. Anaerobic digestion has a different removal rate of ARG host bacteria compared to the other non-host bacteria. Anaerobic digestion also reduced the abundance of integrase genes because most aerobic hosts of integrons cannot withstand the anaerobic environment. The reduction in integrase genes mitigated the horizontal gene transfer of ARGs (Sun et al., 2016).

Both mesophilic (35–37°C) and thermophilic (53–55°C) anaerobic digesters are used for treating organic waste. Compared to mesophilic digestion, thermophilic anaerobic digestion is advantageous in waste stabilization, pathogen elimination, and rapid methane production. However, the operation cost is also higher [158]. The effect of temperature on ARG reduction in anaerobic digestion has been investigated mostly for wastewater sludge [174,179,180] but rarely for dairy manure [181]. Some studies suggest that the thermophilic digestion process [179,181,188] reduces the quantities of mesophilic bacteria that carry certain ARG types and favors the growth of thermophilic bacteria. Thus, the removal of certain ARGs was more efficient compared to the moderate and mesophilic conditions.

However, thermophilic digestion did not always achieve better removal of all ARGs [174,180,189,190]. Ma et al. (2011) conducted laboratory-scale sludge digestion processes, and found that thermophilic anaerobic digestion had a more effective reduction on ermB, ermF, tetO, and tetW, while mesophilic anaerobic digestion significantly reduced sulI, sulII, tetC, tetG, tetX, and intI1. Zhang et al. [174] studied the fate of ARGs through thermophilic and mesophilic anaerobic digestion of wastewater sludge using bench-scale reactors at 35 ± 2°C and 55 ± 2°C, respectively. They found mesophilic anaerobic digestion had more reductions on tetG, tetO, tetW, and ermB, but enriched aadA, macB, and sul1, while thermophilic anaerobic digestion more effectively removed sul2, but enriched erythromycin esterase type I, sul1, and tetM.

Anaerobic digestion can be done in two phases either based on temperature or on the nature of archaea growing in the reactor (acidogenic or methanogenic). Hameed et al. [191] studied anaerobic digestion at two different temperatures (thermophilic and mesophilic). Their findings show that their first phase anaerobic digestion at a thermophilic temperature of 45 and 55°C performed better in terms of methane production and the second phase operated at a mesophilic temperature of 35°C centigrade. They also reported that second phase mesophilic digestion improved the effluent quality by reducing volatile solids by 15–20%. Wu et al. [192] studied the fate of ARGs during two phase anaerobic digestion by metagenomic approach. They divided the anaerobic digestion into acidogenic and methanogenic phases. Their findings show that the acidogenic phase was effective in removing all the ARGs. In our recent study, we found that microwave treatment significantly reduced the ARGs [193]. ARGs such as sulII, intI1, and tnpA were reduced by two-order magnitude in less than 1 min of microwave treatment. Li et al. [194] explored the occurrence of ARGs in sledge samples during microwave-pretreatment and anaerobic digestion. Their results showed that the occurrence of ARGs were quite different. Tian et al. [195] found that the thermophilic anaerobic digestion of sludge under stirring conditions at 55°C led to 55–86% reduction in ARGs. The presence of antibiotic residue did not affect the biogas production.

Recently, Sun et al. [196] showed that biochar has a positive effect on nitrogen conservation during anaerobic digestion process and reduces the abundances of most ARGs in cattle farm wastewater. They found that adding 20 g/L of biochar reduced the rate of ARG spreading. The abundance of 5–7 among the 13 ARGs studied decreased with the addition of biochar. Zhang et al. [197] in their study found that iron nanoparticles not only promote anaerobic digestion and methane production, but also decreased the absolute abundance of total ARGs. Heavy metals have been used as feed additives in livestock feed. Copper is one of the heavy metals that is present in high concentration. Zhang found that in swine manure containing residual copper (75 and 227 mg/L) increased the abundance of ARGs and MGEs. In another study, the same group found that addition of graphene oxide resulted in reduction in ARGs and MEGs abundance [198]. Graphene oxide was found to interfere with the horizontal gene transfer thus limiting the ARG abundance.

3.2.4 Emerging alternate and modified treatment strategies

Electrodialysis reversal was shown to help in the removal of antibiotics, sulfadiazine, and tetracycline [199] from pig manure. The authors found that sulfadiazine was removed mainly due to elector migration and tetracycline was removed by membrane sorption. Li et al. [200] demonstrated that bio-electro-Fenton as a promising system for ARG degradation. They showed that degradation of erythromycin could reach up to 88%. The degradation rate of other genes studied like ermG, ermC, and ermB were 65, 77, and 100%, respectively. Pinecone biochar has been shown to enhance immobilization of SMZ, CIP, and OTC antibiotics [201]. They also found that the kinetics of sorption of these antibiotics were accelerated. Higher sorption to the surfaces led to less runoff of antibiotics.

Quejigo et al. [202] found that using electrode enhances sulfamethazine total bio degradation. In their study, they found that negative anode potential significantly increases antibiotic microbial mineralization. High anode potential enhances the microbial capacity to mineralize sulfamethazine. Li et al. [203] studied the fate of antibiotic-resistant bacteria and genes during electrokinetic treatment of antibiotic-polluted soil. Their findings show that average removal rate of tetracycline, oxytetracycline, and chlortetracycline after 7 days of treatment was 35–40%. They also found that the ARGs tetC, tetG, tetW, and tetM were reduced by an average ratio of 55.5, 12.4, 47.1, and 61.2%, respectively. Ma et al. [204] studied the effect of nano-zerovalent iron on degradation of ARG during anaerobic digestion of cattle manure. Their findings show that 160 mg/L of nano-zerovalent iron effectively removed ARGs. Staley et al. [205] investigated the effect of stockpiling manure on conductive concrete slabs in the removal of antibiotic resistant bacteria and genes. The authors observed a significant reduction in the number of ARGs. A field-based study on farms supplemented antibiotic residues containing manure would be more valuable. Also, it would be important to study the accumulation of antibiotic residues in corn cobs. This would be critical in evaluating the transfer of AR and ARGs if any to humans. Previous studies’ noteworthy findings are reported, when iron-based nanoparticles were used as additives. Iron nanoparticles promoted anaerobic digestion and methane production and decreased the absolute abundance of total ARGs, and the nano-zerovalent iron enhanced the degradation of ARG during anaerobic digestion of cattle manure [225,226]. While the presence of ARGs in manure are reported, how manure borne ARGs are affecting human health is yet to be established, and lack of knowledge in terms of exposure assessment and dose-response assessment data for multiple scenarios is a major challenge [227]. Toxicity data of target antibiotics and non-antibiotics provide comprehensive information about chemical environmental toxicity on aquatic and terrestrial species [228], which can help in approximating lethal concentrations of ARGs. In terms of global health risk, ARGs have accelerated microbial threats to human health in the last decade [229]. Metagenomics-based analysis detected 2,561 ARGs that showed resistance to 24 classes of antibiotics, and about 23.78% of the ARGs pose health risks. Further, extreme climate events such as dry periods followed by rain/flood are shown to increase 2–3 fold higher abundance of ARGs in runoff water [230]. Because of ARGs presence in ambient water flow, most likely ARGs can be transferred from non-organic farms to organic farms. Studies have shown that tetracycline and sulfonamide ARGs were found in soils from organic farms in Nebraska [231]. While ARG/bacteria is currently recognized as a growing problem, quantitative understanding of the direct risk posed by ARGs and antibiotic-resistant bacteria is lacking. Attempts on obtaining the dose-response at lethal and sub-lethal concentrations either through observed studies or mathematical simulation can help in determining the ARGs associated with direct public health risks.

4 Conclusion

The use of antibiotics in animal-agriculture systems is a common practice, and the development of antibiotic resistance acquired by bacteria/pathogens poses risks to animal and human health. Antibiotic resistance is a serious challenge towards the treatment of infectious diseases caused by antibiotic-resistant pathogens. While ARGs are already present in the environment, the excessive use of antibiotics in animal-agriculture systems has the potential to increase the ability of microbial communities to resist the effects of antibiotics. Animal waste borne bacteria are getting increasingly resistant to antibiotics, which could accelerate the emergence and spread of resistance genes in the environment. The study on the abundance of ARGs manure-based farming is a relatively new field of research, and the abundance of ARGs in agricultural soil prior to the application of manure is an important control factor that needs to be taken into account. Animal manure treatment processes such as aerobic digestion, anaerobic digestion, and composting have potential to reduce ARGs in manure. Studies have shown that iron-based addition such as nano-zerovalent iron, and iron nanoparticles promoted anaerobic process, improved methane production, and enhanced ARGs reduction and microbial community. Therefore, the dissemination of these technologies could help in controlling ARGs dissemination. In addition, recently, there has been an effort to find feasible alternatives to heavy use of antibiotics in food animals. While there is ongoing research to identify better alternatives to antibiotics, currently, there is a pressing need to find better ways of treatment to reduce the prevalence of antibiotic residue and ARG in dairy-based manure.

Acknowledgments

We would like to acknowledge the Division of Agriculture and Natural Resources (ANR) and School of Veterinary Medicine Extension, University of California, Davis for supporting this work.

  1. Funding information: The authors state no funding involved.

  2. Author contributions: Y.W. wrote the first draft of manuscript. B.D.S., S.K., and P.K.P. critically revised the manuscript. P.K.P. developed the idea and assisted Y.W. in writing the manuscript. P.K.P. revised the manuscript during review process. All authors read and approved the final manuscript.

  3. Conflict of interest: The authors state no conflict of interest.

  4. Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

[1] CDC. Antibiotic resistance threats in the United States. CDC Washington, DC; 2019. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf Accessed 20 October 2020.Search in Google Scholar

[2] Bush K, Courvalin P, Dantas G, Davies J, Eisenstein B, Huovinen P, et al. Tackling antibiotic resistance. Nat Rev Microbiol. 2011;9(12):894–6. 10.1038/nrmicro2693.Search in Google Scholar PubMed PubMed Central

[3] WHO. Antimicrobial resistance global report on surveillance. World Health Organization; 2014.Search in Google Scholar

[4] Ashbolt NJ, Amézquita A, Backhaus T, Borriello P, Brandt KK, Collignon P, et al. Human health risk assessment (HHRA) for environmental development and transfer of antibiotic resistance. Environ Health Perspect. 2013;121(9):993–1001. 10.1289/ehp.1206316.Search in Google Scholar PubMed PubMed Central

[5] Clardy J, Fischbach MA, Currie CR. The natural history of antibiotics. Curr Biol. 2009;19(11):R437–41. 10.1016/j.cub.2009.04.001.Search in Google Scholar PubMed PubMed Central

[6] Kohanski MA, DePristo MA, Collins JJ. Sublethal antibiotic treatment leads to multidrug resistance via radical-induced mutagenesis. Mol Cell. 2010;37(3):311–20. 10.1016/j.molcel.2010.01.003.Search in Google Scholar PubMed PubMed Central

[7] Ventola CL. The antibiotic resistance crisis: part 1: causes and threats. P T. 2015;40(4):277–83.Search in Google Scholar

[8] Lewis K. Platforms for antibiotic discovery. Nat Rev Drug Discov. 2013;12(5):371–87. 10.1038/nrd3975.Search in Google Scholar PubMed

[9] Berglund B. Environmental dissemination of antibiotic resistance genes and correlation to anthropogenic contamination with antibiotics. Infect Ecol Epidemiol. 2015;5(1):28564. 10.3402/iee.v5.28564.Search in Google Scholar PubMed PubMed Central

[10] Baptista PV, McCusker MP, Carvalho A, Ferreira DA, Mohan NM, Martins M, et al. Nano-strategies to fight multidrug resistant bacteria - “A Battle of the Titans.” Front Microbiol. 2018;9:1441. 10.3389/fmicb.2018.01441.Search in Google Scholar PubMed PubMed Central

[11] Bloom DE, Black S, Salisbury D, Rappuoli R. Antimicrobial resistance and the role of vaccines. Proc Natl Acad Sci. 2018;115(51):12868–71. 10.1073/pnas.1717157115.Search in Google Scholar PubMed PubMed Central

[12] Aslam B, Wang W, Arshad MI, Khurshid M, Muzammil S, Rasool MH, et al. Antibiotic resistance: A rundown of a global crisis. Infect Drug Resist. 2018;11:1645. 10.2147/IDR.S173867.Search in Google Scholar PubMed PubMed Central

[13] O’Neill J. Review on antimicrobial resistance. Antimicrobial resistance: tackling a crisis for the health and wealth of nations 2014. 2014; p. 2–20. https://amr-review.org/sites/default/files/AMR%20Review%20Paper%20-%20Tackling%20a%20crisis%20for%20the%20health%20and%20wealth%20of%20nations_1.pdf (accessed on 9/3/2022).Search in Google Scholar

[14] Martínez JL. Antibiotics and antibiotic resistance genes in natural environments. Science. 2008;321(5887):365–67. 10.1126/science.1159483.Search in Google Scholar PubMed

[15] Ben Y, Fu C, Hu M, Liu L, Wong MH, Zheng C. Human health risk assessment of antibiotic resistance associated with antibiotic residues in the environment: A review. Environ Res. 2019;169:483–93. 10.1016/j.envres.2018.11.040.Search in Google Scholar PubMed

[16] Tangcharoensathien V, Chanvatik S, Sommanustweechai A. Complex determinants of inappropriate use of antibiotics. Bull World Health Organ. 2018;96(2):141. 10.2471/BLT.17.199687. Search in Google Scholar

[17] Magouras I, Carmo LP, Stärk KD, Schüpbach-Regula G. Antimicrobial usage and resistance in livestock: where should we focus? Front veterinary Sci. 2017;4:148. 10.3389/fvets.2017.00148.Search in Google Scholar PubMed PubMed Central

[18] Oliver JP, Gooch CA, Lansing S, Schueler J, Hurst JJ, Sassoubre L, et al. Invited review: Fate of antibiotic residues, antibiotic-resistant bacteria, and antibiotic resistance genes in US dairy manure management systems. J Dairy Sci. 2020;103(2):1051–71. 10.3168/jds.2019-16778.Search in Google Scholar PubMed

[19] Checcucci A, Trevisi P, Luise D, Modesto M, Blasioli S, Braschi I, et al. Exploring the animal waste resistome: the spread of antimicrobial resistance genes through the use of livestock manure. Front Microbiol. 2020;11:1416. 10.3389/fmicb.2020.01416.Search in Google Scholar PubMed PubMed Central

[20] Mirza S, Afzaal M, Begum S, Arooj T, Almas M, Ahmed S. Uptake mechanism of antibiotics in plants. In: Hashmi MZ, editor. Antibiotics and Antimicrobial Resistance Genes in the Environment; 2020. p. 183–188. ISBN: 9780128188828.10.1016/B978-0-12-818882-8.00011-5Search in Google Scholar

[21] Kapoor G, Saigal S, Elongavan A. Action and resistance mechanisms of antibiotics: A guide for clinicians. J Anaesthesiol Clin Pharmacol. 2017;33(3):300–5. 10.4103/joacp.JOACP_349_15.Search in Google Scholar PubMed PubMed Central

[22] O’Rourke A, Beyhan S, Choi Y, Morales P, Chan AP, Espinoza JL, et al. Mechanism-of-action classification of antibiotics by global transcriptome profiling. Antimicrob Agents Chemother. 2020;64(3). 10.1128/AAC.01207-19.Search in Google Scholar PubMed PubMed Central

[23] Ullah H, Ali S. Classification of anti-bacterial agents and their functions. Antibact Agents. 2017;10:1–16. 10.5772/intechopen.68695.Search in Google Scholar

[24] Etebu E, Arikekpar I. Antibiotics: Classification and mechanisms of action with emphasis on molecular perspectives. Int J Appl Microbiol Biotechnol Res. 2016;4:90–101.Search in Google Scholar

[25] Werth BJ. Aminoglycosides, The Merck manual; 2020. https://www.merckmanuals.com/professional/infectious-diseases/bacteria-and-antibacterial-drugs.Search in Google Scholar

[26] Pankey GA, Sabath LD. Clinical relevance of bacteriostatic versus bactericidal mechanisms of action in the treatment of gram-positive bacterial infections. Clin Infect Dis. 2004;38(6):864–70. 10.1086/381972.Search in Google Scholar PubMed

[27] Rahal JJ, Simberkoff MS. Bactericidal and bacteriostatic action of chloramphenicol against meningeal pathogens. Antimicro Agents Chemother. 1979;16(1):13–8. 10.1128/AAC.16.1.13.Search in Google Scholar PubMed PubMed Central

[28] Roberts MC, Schwarz S. Tetracycline and chloramphenicol resistance mechanisms. In: Mayers DL, editor. Antimicrobial drug resistance: infectious disease; 2017. p. 183–193. New York, NY: Humana Press, ISBN: 9783319467160.10.1007/978-1-59745-180-2_15Search in Google Scholar

[29] Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta. 2009;1794(5):808–16. 10.1016/j.bbapap.2008.11.005.Search in Google Scholar PubMed PubMed Central

[30] Vergalli J, Bodrenko IV, Masi M, Moynié L, Acosta-Gutiérrez S, Naismith JH, et al. Porins and small-molecule translocation across the outer membrane of Gram-negative bacteria. Nat Rev Microbiol. 2020;18(3):164–76. 10.1038/s41579-019-0294-2.Search in Google Scholar PubMed

[31] Blair JM, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJ. Molecular mechanisms of antibiotic resistance. Nat Rev Microbiol. 2015;13(1):42–51. 10.1038/nrmicro3380.Search in Google Scholar PubMed

[32] Masi M, Réfregiers M, Pos KM, Pagès J-M. Mechanisms of envelope permeability and antibiotic influx and efflux in Gram-negative bacteria. Nat Microbiol. 2017;2(3):1–7. 10.1038/nmicrobiol.2017.1.Search in Google Scholar PubMed

[33] Fernández L, Hancock REW. Adaptive and mutational resistance: Role of porins and efflux pumps in drug resistance. Clin Microbiol Rev. 2012;25(4):661–81. 10.1128/cmr.00043-12.Search in Google Scholar

[34] Munita JM, Arias CA. Mechanisms of antibiotic resistance. Virulence Mech Bacterial Pathog. 2016;4:481–511. 10.1128/9781555819286.ch17.Search in Google Scholar

[35] Epand RM, Walker C, Epand RF, Magarvey NA. Molecular mechanisms of membrane targeting antibiotics. Biochim Biophys Acta (BBA)-Biomembranes. 2016;1858(5):980–87. 10.1016/j.bbamem.2015.10.018.Search in Google Scholar PubMed

[36] Rouveix B. Clinical implications of multiple drug resistance efflux pumps of pathogenic bacteria. J Antimicrob Chemother. 2007;59(6):1208–9. 10.1093/jac/dkl564.Search in Google Scholar PubMed

[37] Du D, Wang-Kan X, Neuberger A, van Veen HW, Pos KM, Piddock LJ, et al. Multidrug efflux pumps: structure, function and regulation. Nat Rev Microbiol. 2018;16(9):523–39. 10.1038/s41579-018-0048-6.Search in Google Scholar PubMed

[38] Vettoretti L, Plésiat P, Muller C, El Garch F, Phan G, Attrée I, et al. Efflux unbalance in Pseudomonas aeruginosa isolates from cystic fibrosis patients. Antimicrobial Agents Chemotherapy. 2009;53(5):1987–97. 10.1128/AAC.01024-08.Search in Google Scholar PubMed PubMed Central

[39] Alekshun MN, Levy SB. Molecular mechanisms of antibacterial multidrug resistance. Cell. 2007;128(6):1037–50. 10.1016/j.cell.2007.03.004.Search in Google Scholar PubMed

[40] Abraham EP, Chain E. An enzyme from bacteria able to destroy penicillin. Nature. 1940;146(3713):837. 10.1038/146837a0.Search in Google Scholar

[41] Kong K-F, Schneper L, Mathee K. Beta-lactam antibiotics: from antibiosis to resistance and bacteriology. APMIS: Acta Patholog Microbiol Immunol Scand. 2010;118(1):1–36. 10.1111/j.1600-0463.2009.02563.x.Search in Google Scholar PubMed PubMed Central

[42] Bush K, Bradford PA. β-Lactams and β-lactamase inhibitors: An overview. Cold Spring Harb Perspect Med. 2016;6(8):a025247. 10.1101/cshperspect.a025247.Search in Google Scholar PubMed PubMed Central

[43] Christaki E, Marcou M, Tofarides A. Antimicrobial resistance in bacteria: mechanisms, evolution, and persistence. J Mol Evol. 2020;88(1):26–40. 10.1007/s00239-019-09914-3.Search in Google Scholar PubMed

[44] Schaenzer AJ, Wright GD. Antibiotic resistance by enzymatic modification of antibiotic targets. Trends Mol Med. 2020. 10.1016/j.molmed.2020.05.001.Search in Google Scholar PubMed

[45] Cherazard R, Epstein M, Doan T-L, Salim T, Bharti S, Smith MA. Antimicrobial resistant Streptococcus pneumoniae: prevalence, mechanisms, and clinical implications. Am J Therapeutics. 2017;24(3):e361–e69. 10.1097/MJT.0000000000000551.Search in Google Scholar PubMed

[46] Finken M, Kirschner P, Meier A, Wrede A, Böttger EC. Molecular basis of streptomycin resistance in Mycobacterium tuberculosis: alterations of the ribosomal protein S12 gene and point mutations within a functional 16S ribosomal RNA pseudoknot. Mol Microbiol. 1993;9(6):1239–46. 10.1111/j.1365-2958.1993.tb01253.x.Search in Google Scholar PubMed

[47] Olaimat AN, Al-Holy MA, Shahbaz HM, Al-Nabulsi AA, Abu Ghoush MH, Osaili TM, et al. Emergence of antibiotic resistance in Listeria monocytogenes isolated from food products: A comprehensive review. Compr Rev Food Sci Food Saf. 2018;17(5):1277–92. 10.1111/1541-4337.12387.Search in Google Scholar

[48] Enright MC, Robinson DA, Randle G, Feil EJ, Grundmann H, Spratt BG. The evolutionary history of methicillin-resistant Staphylococcus aureus (MRSA). Proc Natl Acad Sci. 2002;99(11):7687–92. 10.1073/pnas.122108599.Search in Google Scholar

[49] Giedraitienė A, Vitkauskienė A, Naginienė R, Pavilonis A. Antibiotic resistance mechanisms of clinically important bacteria. Medicina (Kaunas). 2011;47(3):137–46. 10.3390/medicina47030019.Search in Google Scholar

[50] McGuinness WA, Malachowa N, DeLeo FR. Focus: infectious diseases: vancomycin resistance in Staphylococcus aureus. Yale J Biol Med. 2017;90(2):269.Search in Google Scholar

[51] Pan XS, Fisher LM. Streptococcus pneumoniae DNA gyrase and topoisomerase IV: overexpression, purification, and differential inhibition by fluoroquinolones. Antimicrob Agents Chemother. 1999;43(5):1129–36. 10.1128/AAC.43.5.1129.Search in Google Scholar

[52] Tanaka M, Onodera Y, Uchida Y, Sato K, Hayakawa I. Inhibitory activities of quinolones against DNA gyrase and topoisomerase IV purified from Staphylococcus aureus. Antimicrob Agents Chemother. 1997;41(11):2362–6. 10.1128/AAC.41.11.2362.Search in Google Scholar

[53] Correia S, Poeta P, Hébraud M, Capelo JL, Igrejas G. Mechanisms of quinolone action and resistance: where do we stand. J Med Microbiol. 2017;66(5):551–9. 10.1099/jmm.0.000475.Search in Google Scholar

[54] Telenti A, Imboden P, Marchesi F, Matter L, Schopfer K, Bodmer T, et al. Detection of rifampicin-resistance mutations in Mycobacterium tuberculosis. Lancet. 1993;341(8846):647–51. 10.1016/0140-6736(93)90417-F.Search in Google Scholar

[55] Taniguchi H, Aramaki H, Nikaido Y, Mizuguchi Y, Nakamura M, Koga T, et al. Rifampicin resistance and mutation of the rpoB gene in Mycobacterium tuberculosis. FEMS Microbiol Lett. 1996;144(1):103–08. 10.1111/j.1574-6968.1996.tb08515.x.Search in Google Scholar PubMed

[56] Singh A, Grover S, Sinha S, Das M, Somvanshi P, Grover A. Mechanistic principles behind molecular mechanism of rifampicin resistance in mutant RNA polymerase beta subunit of Mycobacterium tuberculosis. J Cell Biochem. 2017;118(12):4594–606. 10.1002/jcb.26124.Search in Google Scholar PubMed

[57] Kirchhelle C. Pharming animals: a global history of antibiotics in food production (1935–2017). Palgrave Commun. 2018;4(1):1–13. 10.1057/s41599-018-0152-2.Search in Google Scholar

[58] FFSA. In the Finnish food safety authority E, and the faculty of veterinary medicine at the university of Helsinki, Ed; 2018.Search in Google Scholar

[59] Ibrahim M, Ahmad F, Yaqub B, Ramzan A, Imran A, Afzaal M, et al. Current trends of antimicrobials used in food animals and aquaculture. Antibiotics Antimicrob Resist Genes Environ. 2020;1:39–69. 10.1016/B978-0-12-818882-8.00004-8.Search in Google Scholar

[60] De Briyne N, Atkinson J, Pokludová L, Borriello S. Antibiotics used most commonly to treat animals in Europe. Veterinary Rec. 2014;175(13):325. 10.1136/vr.102462.Search in Google Scholar PubMed PubMed Central

[61] Van Boeckel TP, Brower C, Gilbert M, Grenfell BT, Levin SA, Robinson TP, et al. Global trends in antimicrobial use in food animals. Proc Natl Acad Sci. 2015;112(18):5649–54. 10.1073/pnas.1503141112.Search in Google Scholar PubMed PubMed Central

[62] FDA. Summary report on antimicrobials sold or distributed for use in food-producing animals; 2020. https://www.fda.gov/media/144427/download. Accessed 31 December 2020.Search in Google Scholar

[63] FAO. Food outlook: Biannual report on global food markets. Food and Agriculture Organization of the United Nations; 2020. http://www.fao.org/documents/card/en/c/ca9509en/. Accessed 20 October 2020.Search in Google Scholar

[64] NASS. 2012 Census of Agriculture. United States summary and state data. Vol. 1. Geographic area series part 51. USDA, National Agricultural Statistics Service, AC-12-A-51; 2014. https://www.nass.usda.gov/Publications/AgCensus/2012/index.php. Accessed 20 October 2020.Search in Google Scholar

[65] NAHMS. Dairy 2014 health and management practices in US Dairy operation, 2014. USDA, Animal and Plant Health Inspection Service, Veterinary Services, Center for Epidemiology and Animal Health, National Animal Health Monitoring System; 2018. https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy14/Dairy14_dr_PartIII.pdf. Accessed 20 October 2020.Search in Google Scholar

[66] Landers TF, Cohen B, Wittum TE, Larson EL. A review of antibiotic use in food animals: perspective, policy, and potential. Public Health Rep. 2012;127(1):4–22. 10.1177/003335491212700103.Search in Google Scholar PubMed PubMed Central

[67] USDA. Reference of dairy cattle health and management practices in the United States. US Department of Agriculture, National Animal Health Monitoring System; 2007. https://www.aphis.usda.gov/animal_health/nahms/dairy/downloads/dairy07/Dairy07_allpubs.pdf. Accessed 20 October 2020.Search in Google Scholar

[68] Dargatz D. Feedlot 99, Part Iii: health management and biosecurity in US feedlots, 1999; 2000.Search in Google Scholar

[69] Word AB, Wickersham TA, Trubenbach LA, Mays GB, Sawyer JE. Effects of metaphylaxis on production responses and total antimicrobial use in high-risk beef calves. Appl Anim Sci. 2020;36(2):265–70. 10.15232/aas.2019-01914.Search in Google Scholar

[70] Bogueva D, Marinova D, Raphaely T, editors. Handbook of research on social marketing and its influence on animal origin food product consumption. IGI Global. 2018; p. 77–91. 10.4018/978-1-5225-4757-0.Search in Google Scholar

[71] Diana A, Boyle LA, Leonard FC, Carroll C, Sheehan E, Murphy D, et al. Removing prophylactic antibiotics from pig feed: how does it affect their performance and health? BMC Vet Res. 2019;15(1):67. 10.1186/s12917-019-1808-x.Search in Google Scholar PubMed PubMed Central

[72] Lorenz I, Fagan J, More SJ. Calf health from birth to weaning. II. Management of diarrhoea in pre-weaned calves. Ir Veterinary J. 2011;64(1):9. 10.1186/2046-0481-64-9.Search in Google Scholar PubMed PubMed Central

[73] Agga GE, Schmidt JW, Arthur TM. Effects of in-feed chlortetracycline prophylaxis in beef cattle on animal health and antimicrobial-resistant Escherichia coli. Appl Environ Microbiol. 2016;82(24):7197–204. 10.1128/AEM.01928-16.Search in Google Scholar PubMed PubMed Central

[74] Kertz A, Hill T, Quigley J, III, Heinrichs A, Linn J, Drackley J. A 100-Year Review: Calf nutrition and management. J Dairy Sci. 2017;100(12):10151–72. 10.3168/jds.2017-13062.Search in Google Scholar PubMed

[75] Ronquillo MG, Hernandez JCA. Antibiotic and synthetic growth promoters in animal diets: Review of impact and analytical methods. Food Control. 2017;72:255–67. 10.1016/j.foodcont.2016.03.001.Search in Google Scholar

[76] Viola C, DeVincent SJ. Overview of issues pertaining to the manufacture, distribution, and use of antimicrobials in animals and other information relevant to animal antimicrobial use data collection in the United States. Prev Vet Med. 2006;73(2–3):111–31. 10.1016/j.prevetmed.2005.09.020.Search in Google Scholar

[77] Dibner JJ, Richards JD. Antibiotic growth promoters in agriculture: history and mode of action. Poult Sci. 2005;84(4):634–43. 10.1093/ps/84.4.634.Search in Google Scholar

[78] Holman DB, Chénier MR. Antimicrobial use in swine production and its effect on the swine gut microbiota and antimicrobial resistance. Can J Microbiol. 2015;61(11):785–98. 10.1139/cjm-2015-0239.Search in Google Scholar

[79] Butaye P, Devriese LA, Haesebrouck F. Antimicrobial growth promoters used in animal feed: effects of less well known antibiotics on gram-positive bacteria. Clin Microbiol Rev. 2003;16(2):175–88. 10.1128/CMR.16.2.175-188.2003.Search in Google Scholar

[80] Cromwell GL. Why and how antibiotics are used in swine production. Anim Biotechnol. 2002;13(1):7–27. 10.1081/ABIO-120005767.Search in Google Scholar

[81] Brown K, Uwiera RR, Kalmokoff ML, Brooks SP, Inglis GD. Antimicrobial growth promoter use in livestock: A requirement to understand their modes of action to develop effective alternatives. Int J Antimicrob Agents. 2017;49(1):12–24. 10.1016/j.ijantimicag.2016.08.006.Search in Google Scholar

[82] Tang KL, Caffrey NP, Nóbrega DB, Cork SC, Ronksley PE, Barkema HW, 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(8):e316–e27. 10.1016/S2542-5196(17)30141-9.Search in Google Scholar

[83] Scott AM, Beller E, Glasziou P, Clark J, Ranakusuma RW, Byambasuren O, et al. Is antimicrobial administration to food animals a direct threat to human health? A rapid systematic review. Int J Antimicrob Agents. 2018;52(3):316–23. 10.1016/j.ijantimicag.2018.04.005.Search in Google Scholar PubMed

[84] Smith-Spangler C, Brandeau ML, Hunter GE, Bavinger JC, Pearson M, Eschbach PJ, et al. Are organic foods safer or healthier than conventional alternatives? A systematic review. Ann Intern Med. 2012;157(5):348–66. 10.7326/0003-4819-157-5-201209040-00007.Search in Google Scholar PubMed

[85] McEwen SA, Collignon PJ. Antimicrobial resistance: A one health perspective. Microbiol Spectr. 2018;6(2):6–12. 10.1128/9781555819804.ch25.Search in Google Scholar

[86] Levison ME, Levison JH. Pharmacokinetics and pharmacodynamics of antibacterial agents. Infect Dis Clin North Am. 2009;23(4):791–815. 10.1016/j.idc.2009.06.008.Search in Google Scholar PubMed PubMed Central

[87] Sarmah AK, Meyer MT, Boxall AB. A global perspective on the use, sales, exposure pathways, occurrence, fate and effects of veterinary antibiotics (VAs) in the environment. Chemosphere. 2006;65(5):725–59. 10.1016/j.chemosphere.2006.03.026.Search in Google Scholar PubMed

[88] Wallace JS, Aga DS. Enhancing extraction and detection of veterinary antibiotics in solid and liquid fractions of manure. J Environ Qual. 2016;45(2):471–9. 10.2134/jeq2015.05.0246.Search in Google Scholar PubMed

[89] Looft T, Johnson TA, Allen HK, Bayles DO, Alt DP, Stedtfeld RD, et al. In-feed antibiotic effects on the swine intestinal microbiome. Proc Natl Acad Sci. 2012;109(5):1691–6. 10.1073/pnas.1120238109.Search in Google Scholar PubMed PubMed Central

[90] Jeters RT, Wang G-R, Moon K, Shoemaker NB, Salyers AA. Tetracycline-associated transcriptional regulation of transfer genes of the Bacteroides conjugative transposon CTnDOT. J Bacteriol. 2009;191(20):6374–82. 10.1128/JB.00739-09.Search in Google Scholar PubMed PubMed Central

[91] Pereira R, Siler J, Ng J, Davis M, Grohn Y, Warnick L. Effect of on-farm use of antimicrobial drugs on resistance in fecal Escherichia coli of preweaned dairy calves. J Dairy Sci. 2014;97(12):7644–54. 10.3168/jds.2014-8521.Search in Google Scholar PubMed PubMed Central

[92] Qian X, Gu J, Sun W, Wang X-J, Su J-Q, Stedfeld R. Diversity, abundance, and persistence of antibiotic resistance genes in various types of animal manure following industrial composting. J Hazard Mater. 2018;344:716–22. 10.1016/j.jhazmat.2017.11.020.Search in Google Scholar PubMed

[93] McKinney CW, Dungan RS, Moore A, Leytem AB. Occurrence and abundance of antibiotic resistance genes in agricultural soil receiving dairy manure. FEMS Microbiol Ecol. 2018;94(3):fiy010 10.1093/femsec/fiy010.Search in Google Scholar PubMed

[94] Yuan QB, Zhai YF, Mao BY, Schwarz C, Hu N. Fates of antibiotic resistance genes in a distributed swine wastewater treatment plant. Water Environ Res. 2019;91(12):1565–75. 10.1002/wer.1125.Search in Google Scholar PubMed

[95] He Y, Yuan Q, Mathieu J, Stadler L, Senehi N, Sun R, et al. Antibiotic resistance genes from livestock waste: occurrence, dissemination, and treatment. NPJ Clean Water. 2020;3(1):4. 10.1038/s41545-020-0051-0.Search in Google Scholar

[96] Zhu Y-G, Johnson TA, Su J-Q, Qiao M, Guo G-X, Stedtfeld RD, et al. Diverse and abundant antibiotic resistance genes in Chinese swine farms. Proc Natl Acad Sci. 2013;110(9):3435–40. 10.1073/pnas.1222743110.Search in Google Scholar PubMed PubMed Central

[97] Gou M, Hu H-W, Zhang Y-J, Wang J-T, Hayden H, Tang Y-Q, et al. Aerobic composting reduces antibiotic resistance genes in cattle manure and the resistome dissemination in agricultural soils. Sci Total Environ. 2018;612:1300–10. 10.1016/j.scitotenv.2017.09.028.Search in Google Scholar PubMed

[98] Tien Y-C, Li B, Zhang T, Scott A, Murray R, Sabourin L, et al. Impact of dairy manure pre-application treatment on manure composition, soil dynamics of antibiotic resistance genes, and abundance of antibiotic-resistance genes on vegetables at harvest. Sci Total Environ. 2017;581:32–9. 10.1016/j.scitotenv.2016.12.138.Search in Google Scholar PubMed

[99] Awasthi MK, Liu T, Chen H, Verma S, Duan Y, Awasthi SK, et al. The behavior of antibiotic resistance genes and their associations with bacterial community during poultry manure composting. Bioresour Technol. 2019;280:70–8. 10.1016/j.biortech.2019.02.030.Search in Google Scholar PubMed

[100] Zhang R, Wang X, Gu J, Zhang Y. Influence of zinc on biogas production and antibiotic resistance gene profiles during anaerobic digestion of swine manure. Bioresour Technol. 2017;244:63–70. 10.1016/j.biortech.2017.07.032.Search in Google Scholar PubMed

[101] Werner G, Klare I, Heier H, Hinz K-H, Böhme G, Wendt M, et al. Quinupristin/dalfopristin-resistant enterococci of the satA (vatD) and satG (vatE) genotypes from different ecological origins in Germany. Microb Drug Resist. 2000;6(1):37–47. 10.1089/mdr.2000.6.37.Search in Google Scholar PubMed

[102] Cui E, Wu Y, Zuo Y, Chen H. Effect of different biochars on antibiotic resistance genes and bacterial community during chicken manure composting. Bioresour Technol. 2016;203:11–7. 10.1016/j.biortech.2015.12.030.Search in Google Scholar PubMed

[103] Alanis AJ. Resistance to antibiotics: are we in the post-antibiotic era? Arch Med Res. 2005;36(6):697–705. 10.1016/j.arcmed.2005.06.009.Search in Google Scholar PubMed

[104] Wozniak RA, Waldor MK. Integrative and conjugative elements: mosaic mobile genetic elements enabling dynamic lateral gene flow. Nat Rev Microbiol. 2010;8(8):552–63. 10.1038/nrmicro2382.Search in Google Scholar PubMed

[105] Penadés JR, Chen J, Quiles-Puchalt N, Carpena N, Novick RP. Bacteriophage-mediated spread of bacterial virulence genes. Curr Opin Microbiol. 2015;23:171–8. 10.1016/j.mib.2014.11.019.Search in Google Scholar PubMed

[106] Seitz P, Blokesch M. Cues and regulatory pathways involved in natural competence and transformation in pathogenic and environmental Gram-negative bacteria. FEMS Microbiol Rev. 2013;37(3):336–63. 10.1111/j.1574-6976.2012.00353.x.Search in Google Scholar PubMed

[107] Lerminiaux NA, Cameron AD. Horizontal transfer of antibiotic resistance genes in clinical environments. Can J Microbiol. 2019;65(1):34–44. 10.1139/cjm-2018-0275.Search in Google Scholar PubMed

[108] Torres-Barceló C. The disparate effects of bacteriophages on antibiotic-resistant bacteria. Emerg Microbes Infect. 2018;7(1):1–12. 10.1038/s41426-018-0169-z.Search in Google Scholar PubMed PubMed Central

[109] Ross J, Topp E. Abundance of antibiotic resistance genes in bacteriophage following soil fertilization with dairy manure or municipal biosolids, and evidence for potential transduction. Appl Environ Microbiol. 2015;81(22):7905–13. 10.1128/AEM.02363-15.Search in Google Scholar PubMed PubMed Central

[110] Zhang P-Y, Xu P-P, Xia Z-J, Wang J, Xiong J, Li Y-Z. Combined treatment with the antibiotics kanamycin and streptomycin promotes the conjugation of Escherichia coli. FEMS Microbiol Lett. 2013;348(2):149–56. 10.1111/1574-6968.12282.Search in Google Scholar PubMed

[111] Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev. 2010;74(3):417–33. 10.1128/MMBR.00016-10.Search in Google Scholar PubMed PubMed Central

[112] Yan C, Yang Y, Zhou J, Liu M, Nie M, Shi H, et al. Antibiotics in the surface water of the Yangtze Estuary: Occurrence, distribution and risk assessment. Environ Pollut. 2013;175:22–9. 10.1016/j.envpol.2012.12.008.Search in Google Scholar PubMed

[113] Vikesland PJ, Pruden A, Alvarez PJ, Aga D, Bürgmann H, Li X-D, et al. Toward a comprehensive strategy to mitigate dissemination of environmental sources of antibiotic resistance. Environ. Sci. Technol. 2017;51:13061–9. https://doi.org/10.1021/acs.est.7b03623.10.1021/acs.est.7b03623Search in Google Scholar PubMed

[114] Johnson TA, Stedtfeld RD, Wang Q, Cole JR, Hashsham SA, Looft T, et al. Clusters of antibiotic resistance genes enriched together stay together in swine agriculture. MBio. 2016;7(2):e02214–15. 10.1128/mBio.02214-15.Search in Google Scholar PubMed PubMed Central

[115] Han X-M, Hu H-W, Chen Q-L, Yang L-Y, Li H-L, Zhu Y-G, et al. Antibiotic resistance genes and associated bacterial communities in agricultural soils amended with different sources of animal manures. Soil Biol Biochem. 2018;126:91–102. 10.1016/j.soilbio.2018.08.018.Search in Google Scholar

[116] Williams‐Nguyen J, Sallach JB, Bartelt‐Hunt S, Boxall AB, Durso LM, McLain JE, et al. Antibiotics and antibiotic resistance in agroecosystems: state of the science. J Environ Qual. 2016;45(2):394–406. 10.2134/jeq2015.07.0336.Search in Google Scholar PubMed

[117] Chee-Sanford JC, Mackie RI, Koike S, Krapac IG, Lin Y-F, Yannarell AC, et al. Fate and transport of antibiotic residues and antibiotic resistance genes following land application of manure waste. J Environ Qual. 2009;38(3):1086–108. 10.2134/jeq2008.0128.Search in Google Scholar PubMed

[118] Dong P, Wang H, Fang T, Wang Y, Ye Q. Assessment of extracellular antibiotic resistance genes (eARGs) in typical environmental samples and the transforming ability of eARG. Environ Int. 2019;125:90–6. 10.1016/j.envint.2019.01.050.Search in Google Scholar PubMed

[119] Davies J, Spiegelman GB, Yim G. The world of subinhibitory antibiotic concentrations. Curr Opin Microbiol. 2006;9(5):445–53. 10.1016/j.mib.2006.08.006.Search in Google Scholar PubMed

[120] Poole K. Bacterial stress responses as determinants of antimicrobial resistance. J Antimicrob Chemother. 2012;67(9):2069–89. 10.1093/jac/dks196.Search in Google Scholar PubMed

[121] Beaber JW, Hochhut B, Waldor MK. SOS response promotes horizontal dissemination of antibiotic resistance genes. Nature. 2004;427(6969):72–4. 10.1038/nature02241.Search in Google Scholar PubMed

[122] VanBogelen RA, Neidhardt FC. Ribosomes as sensors of heat and cold shock in Escherichia coli. Proc Natl Acad Sci. 1990;87(15):5589–93. 10.1073/pnas.87.15.5589.Search in Google Scholar PubMed PubMed Central

[123] Pruden A, Larsson DJ, Amézquita A, Collignon P, Brandt KK, Graham DW, et al. Management options for reducing the release of antibiotics and antibiotic resistance genes to the environment. Environ Health Perspect. 2013;121(8):878. 10.1289/ehp.1206446.Search in Google Scholar PubMed PubMed Central

[124] Baker-Austin C, Wright MS, Stepanauskas R, McArthur J. Co-selection of antibiotic and metal resistance. Trends Microbiol. 2006;14(4):176–82. 10.1016/j.tim.2006.02.006.Search in Google Scholar PubMed

[125] Ghosh S, LaPara TM. The effects of subtherapeutic antibiotic use in farm animals on the proliferation and persistence of antibiotic resistance among soil bacteria. ISME J. 2007;1(3):191–203. 10.1038/ismej.2007.31.Search in Google Scholar PubMed

[126] Marti R, Scott A, Tien Y-C, Murray R, Sabourin L, Zhang Y, et al. Impact of manure fertilization on the abundance of antibiotic-resistant bacteria and frequency of detection of antibiotic resistance genes in soil and on vegetables at harvest. Appl Environ Microbiol. 2013;79(18):5701–9. 10.1128/AEM.01682-13.Search in Google Scholar PubMed PubMed Central

[127] Chee-Sanford JC, Aminov RI, Krapac IJ, Garrigues-Jeanjean N, Mackie RI. Occurrence and diversity of tetracycline resistance genes in lagoons and groundwater underlying two swine production facilities. Appl Environ Microbiol. 2001;67(4):1494–502. 10.1128/aem.67.4.1494-1502.2001.Search in Google Scholar

[128] Koike S, Krapac I, Oliver H, Yannarell A, Chee-Sanford J, Aminov R, et al. Monitoring and source tracking of tetracycline resistance genes in lagoons and groundwater adjacent to swine production facilities over a 3-year period. Appl Environ Microbiol. 2007;73(15):4813–23. 10.1128/AEM.00665-07.Search in Google Scholar PubMed PubMed Central

[129] Allen HK, Donato J, Wang HH, Cloud-Hansen KA, Davies J, Handelsman J. Call of the wild: antibiotic resistance genes in natural environments. Nat Rev Microbiol. 2010;8(4):251–9. 10.1038/nrmicro2312.Search in Google Scholar PubMed

[130] Heo Y, Park J, Lim SI, Hur HG, Kim D, Park K. Size-resolved culturable airborne bacteria sampled in rice field, sanitary landfill, and waste incineration sites. J Env Monit. 2010;12(8):1619–24. 10.1039/c0em00004c.Search in Google Scholar PubMed

[131] Létourneau V, Nehmé B, Mériaux A, Massé D, Cormier Y, Duchaine C. Human pathogens and tetracycline-resistant bacteria in bioaerosols of swine confinement buildings and in nasal flora of hog producers. Int J Hyg Environ Health. 2010;213(6):444–49. 10.1016/j.ijheh.2010.09.008.Search in Google Scholar PubMed

[132] Rule AM, Evans SL, Silbergeld EK. Food animal transport: A potential source of community exposures to health hazards from industrial farming (CAFOs. J Infect Public Health. 2008;1(1):33–9. 10.1016/j.jiph.2008.08.001.Search in Google Scholar PubMed

[133] Forsberg KJ, Reyes A, Wang B, Selleck EM, Sommer MO, Dantas G. The shared antibiotic resistome of soil bacteria and human pathogens. Science. 2012;337(6098):1107–11. 10.1126/science.1220761.Search in Google Scholar PubMed PubMed Central

[134] Reynaga E, Navarro M, Vilamala A, Roure P, Quintana M, Garcia-Nuñez M, et al. Prevalence of colonization by methicillin-resistant Staphylococcus aureus ST398 in pigs and pig farm workers in an area of Catalonia, Spain. BMC Infect Dis. 2016;16(1):716. 10.1186/s12879-016-2050-9.Search in Google Scholar PubMed PubMed Central

[135] Hatcher SM, Rhodes SM, Stewart JR, Silbergeld E, Pisanic N, Larsen J, et al. The prevalence of antibiotic-resistant Staphylococcus aureus nasal carriage among industrial hog operation workers, community residents, and children living in their households: North Carolina, USA. Environ Health Perspect. 2017;125(4):560–9. 10.1289/EHP35.Search in Google Scholar

[136] Johnsen PJ, Townsend JP, Bøhn T, Simonsen GS, Sundsfjord A, Nielsen KM. Factors affecting the reversal of antimicrobial-drug resistance. Lancet Infect Dis. 2009;9(6):357–64. 10.1016/S1473-3099(09)70105-7.Search in Google Scholar

[137] Hill D, Morra MJ, Stalder T, Jechalke S, Top E, Pollard AT, et al. Dairy manure as a potential source of crop nutrients and environmental contaminants. J Environ Sci. 2021;100:117–30. 10.1016/j.jes.2020.07.016.Search in Google Scholar PubMed

[138] Ruuskanen M, Muurinen J, Meierjohan A, Pärnänen K, Tamminen M, Lyra C, et al. Fertilizing with animal manure disseminates antibiotic resistance genes to the farm environment. J Environ Qual. 2016;45(2):488–93. 10.2134/jeq2015.05.0250.Search in Google Scholar PubMed

[139] Gao M, Qiu T, Sun Y, Wang X. The abundance and diversity of antibiotic resistance genes in the atmospheric environment of composting plants. Environ Int. 2018;116:229–38. 10.1016/j.envint.2018.04.028.Search in Google Scholar PubMed

[140] Guron GK, Arango-Argoty G, Zhang L, Pruden A, Ponder MA. Effects of dairy manure-based amendments and soil texture on lettuce-and radish-associated microbiota and resistomes. Msphere. 2019;4(3):e00239–19. 10.1128/mSphere.00239-19.Search in Google Scholar PubMed PubMed Central

[141] Zhang Y-J, Hu H-W, Chen Q-L, Singh BK, Yan H, Chen D, et al. Transfer of antibiotic resistance from manure-amended soils to vegetable microbiomes. Environ Int. 2019;130:104912. 10.1016/j.envint.2019.104912.Search in Google Scholar PubMed

[142] Zhang Y-J, Hu H-W, Chen Q-L, Yan H, Wang J-T, Chen D, et al. Manure application did not enrich antibiotic resistance genes in root endophytic bacterial microbiota of cherry radish plants. Appl Environ Microbiol. 2020;86(2):e02106–19. 10.1128/AEM.02106-19.Search in Google Scholar PubMed PubMed Central

[143] Mullen RA, Hurst JJ, Naas KM, Sassoubre LM, Aga DS. Assessing uptake of antimicrobials by Zea mays L. and prevalence of antimicrobial resistance genes in manure-fertilized soil. Sci Total Environ. 2019;646:409–15. 10.1016/j.scitotenv.2018.07.199.Search in Google Scholar PubMed

[144] Zhu D, Giles M, Yang X-R, Daniell T, Neilson R, Zhu Y-G. Phyllosphere of staple crops under pig manure fertilization, a reservoir of antibiotic resistance genes. Environ Pollut. 2019;252:227–35. 10.1016/j.envpol.2019.05.098.Search in Google Scholar PubMed

[145] Zhao F, Yang L, Chen L, Li S, Sun L. Bioaccumulation of antibiotics in crops under long-term manure application: Occurrence, biomass response and human exposure. Chemosphere. 2019;219:882–95. 10.1016/j.chemosphere.2018.12.076.Search in Google Scholar PubMed

[146] FDA. Food/Guidance & Regulation (Food and Dietary Supplements)/Food Safety Modernization Act (FSMA)/Raw Manure under the FSMA Final Rule on Produce Safety; 2018. https://www.fda.gov/food/food-safety-modernization-act-fsma/raw-manure-under-fsma-final-rule-produce-safety.Search in Google Scholar

[147] Garcia M, Szogi A, Vanotti M, Chastain J, Millner P. Enhanced solid–liquid separation of dairy manure with natural flocculants. Bioresour Technol. 2009;100(22):5417–23. 10.1016/j.biortech.2008.11.012.Search in Google Scholar PubMed

[148] Hurst JJ, Oliver JP, Schueler J, Gooch C, Lansing S, Crossette E, et al. Trends in antimicrobial resistance genes in manure blend pits and long-term storage across dairy farms with comparisons to antimicrobial usage and residual concentrations. Environ Sci Technol. 2019;53(5):2405–15. 10.1021/acs.est.8b05702.Search in Google Scholar PubMed

[149] Wallace JS, Garner E, Pruden A, Aga DS. Occurrence and transformation of veterinary antibiotics and antibiotic resistance genes in dairy manure treated by advanced anaerobic digestion and conventional treatment methods. Environ Pollut. 2018;236:764–72. 10.1016/j.envpol.2018.02.024.Search in Google Scholar PubMed

[150] Gros M, Marti E, Balcázar JL, Boy-Roura M, Busquets A, Colon J, et al. Fate of pharmaceuticals and antibiotic resistance genes in a full-scale on-farm livestock waste treatment plant. J Hazard Mater. 2019;378:120716. 10.1016/j.jhazmat.2019.05.109.Search in Google Scholar PubMed

[151] Zhang Q, Xu J, Wang X, Zhu W, Pang X, Zhao J. Changes and distributions of antibiotic resistance genes in liquid and solid fractions in mesophilic and thermophilic anaerobic digestion of dairy manure. Bioresour Technol. 2021;320:124372. 10.1016/j.biortech.2020.124372.Search in Google Scholar PubMed

[152] Wright P, Gooch C. Antibiotic residuals and solid-liquid separation of dairy manure; 2020;1–2. Dairy Environmental Systems Program. https://ecommons.cornell.edu/handle/1813/102697. (accessed on 9/2/2022).Search in Google Scholar

[153] Su J-Q, Wei B, Ou-Yang W-Y, Huang F-Y, Zhao Y, Xu H-J, et al. Antibiotic resistome and its association with bacterial communities during sewage sludge composting. Environ. Sci. Technol. 2015;49(12):7356–63. 10.1021/acs.est.5b01012.Search in Google Scholar PubMed

[154] Wang J, Ben W, Zhang Y, Yang M, Qiang Z. Effects of thermophilic composting on oxytetracycline, sulfamethazine, and their corresponding resistance genes in swine manure. Environ Sci Process Impacts. 2015;17(9):1654–60. 10.1039/C5EM00132C.Search in Google Scholar PubMed

[155] Chen J, Yu Z, Michel FC, Wittum T, Morrison M. Development and application of real-time PCR assays for quantification of erm genes conferring resistance to macrolides-lincosamides-streptogramin B in livestock manure and manure management systems. Appl Environ Microbiol. 2007;73(14):4407–16. 10.1128/AEM.02799-06.Search in Google Scholar PubMed PubMed Central

[156] Wang L, Oda Y, Grewal S, Morrison M, Michel FC, Yu Z. Persistence of resistance to erythromycin and tetracycline in swine manure during simulated composting and lagoon treatments. Microb Ecol. 2012;63(1):32–40. 10.1007/s00248-011-9921-9.Search in Google Scholar PubMed

[157] Selvam A, Xu D, Zhao Z, Wong JW. Fate of tetracycline, sulfonamide and fluoroquinolone resistance genes and the changes in bacterial diversity during composting of swine manure. Bioresour Technol. 2012;126:383–90. 10.1016/j.biortech.2012.03.045.Search in Google Scholar PubMed

[158] Youngquist CP, Mitchell SM, Cogger CG. Fate of antibiotics and antibiotic resistance during digestion and composting: A review. J Environ Qual. 2016;45(2):537–45. 10.2134/jeq2015.05.0256.Search in Google Scholar PubMed

[159] Xu S, Sura S, Zaheer R, Wang G, Smith A, Cook S, et al. Dissipation of antimicrobial resistance determinants in composted and stockpiled beef cattle manure. J Environ Qual. 2016;45(2):528–36. 10.2134/jeq2015.03.0146.Search in Google Scholar PubMed

[160] Xie WY, Yang XP, Li Q, Wu LH, Shen QR, Zhao FJ. Changes in antibiotic concentrations and antibiotic resistome during commercial composting of animal manures. Env Pollut. 2016;219:182–90. 10.1016/j.envpol.2016.10.044.Search in Google Scholar PubMed

[161] Qian X, Sun W, Gu J, Wang X-J, Zhang Y-J, Duan M-L, et al. Reducing antibiotic resistance genes, integrons, and pathogens in dairy manure by continuous thermophilic composting. Bioresour Technol. 2016;220:425–32. 10.1016/j.biortech.2016.08.101.Search in Google Scholar PubMed

[162] Awasthi MK, Chen H, Awasthi SK, Duan Y, Liu T, Pandey A, et al. Application of metagenomic analysis for detection of the reduction in the antibiotic resistance genes (ARGs) by the addition of clay during poultry manure composting. Chemosphere. 2019;220:137–45. 10.1016/j.chemosphere.2018.12.103.Search in Google Scholar PubMed

[163] Guo A, Gu J, Wang X, Zhang R, Yin Y, Sun W, et al. Effects of superabsorbent polymers on the abundances of antibiotic resistance genes, mobile genetic elements, and the bacterial community during swine manure composting. Bioresour Technol. 2017;244:658–63. 10.1016/j.biortech.2017.08.016.Search in Google Scholar PubMed

[164] Peng S, Li H, Song D, Lin X, Wang Y. Influence of zeolite and superphosphate as additives on antibiotic resistance genes and bacterial communities during factory-scale chicken manure composting. Bioresour Technol. 2018;263:393–401. 10.1016/j.biortech.2018.04.107.Search in Google Scholar PubMed

[165] Liao H, Lu X, Rensing C, Friman VP, Geisen S, Chen Z, et al. Hyperthermophilic composting accelerates the removal of antibiotic resistance genes and mobile genetic elements in sewage sludge. Environ Sci Technol. 2018;52(1):266–76. 10.1021/acs.est.7b04483.Search in Google Scholar PubMed

[166] Cao R, Wang J, Ben W, Qiang Z. The profile of antibiotic resistance genes in pig manure composting shaped by composting stage: Mesophilic-thermophilic and cooling-maturation stages. Chemosphere. 2020;250:126181. 10.1016/j.chemosphere.2020.126181.Search in Google Scholar PubMed

[167] Cao R, Ben W, Qiang Z, Zhang J. Removal of antibiotic resistance genes in pig manure composting influenced by inoculation of compound microbial agents. Bioresour Technol. 2020;317:123966. 10.1016/j.biortech.2020.123966.Search in Google Scholar PubMed

[168] Li H, Zheng X, Cao H, Tan L, Yang B, Cheng W, et al. Reduction of antibiotic resistance genes under different conditions during composting process of aerobic combined with anaerobic. Bioresour Technol. 2021;325:124710. 10.1016/j.biortech.2021.124710.Search in Google Scholar PubMed

[169] Yu H, Zhao Q, Meng F, Ruan L, Sun T, Liu X, et al. Deciphering the role of calcium peroxide on the fate of antibiotic resistance genes and mobile genetic elements during bioelectrochemically-assisted anaerobic composting of excess dewatered sludge. Chem Eng J. 2020;397:125355. 10.1016/j.cej.2020.125355.Search in Google Scholar

[170] Li H, Cheng W, Li B, Xu Y, Zheng X. The fate of antibiotic resistance genes during co-composting of swine manure with cauliflower and corn straw. Bioresour Technol. 2020;300:122669. 10.1016/j.biortech.2019.122669.Search in Google Scholar PubMed

[171] Tang Z, Xi B, Huang C, Tan W, Li W, Zhao X, et al. Mobile genetic elements in potential host microorganisms are the key hindrance for the removal of antibiotic resistance genes in industrial-scale composting with municipal solid waste. Bioresour Technol. 2020;301:122723. 10.1016/j.biortech.2019.122723.Search in Google Scholar PubMed

[172] Zhang L, Li L, Sha G, Liu C, Wang Z, Wang L. Aerobic composting as an effective cow manure management strategy for reducing the dissemination of antibiotic resistance genes: An integrated meta-omics study. J Hazard Mater. 2020;386:121895. 10.1016/j.jhazmat.2019.121895.Search in Google Scholar PubMed

[173] Flores Orozco D. Effect of ceftiofur on mesophilic anaerobic digestion of dairy manure and the removal of the cephalosporin-resistance gene CMY-2. Bioresour Technol. 2020;301:122729. 10.1016/j.biortech.2019.122729.Search in Google Scholar PubMed

[174] Zhang T, Yang Y, Pruden A. Effect of temperature on removal of antibiotic resistance genes by anaerobic digestion of activated sludge revealed by metagenomic approach. Appl Microbiol Biotechnol. 2015;99(18):7771–9. 10.1007/s00253-015-6688-9.Search in Google Scholar PubMed

[175] Song W, Wang X, Gu J, Zhang S, Yin Y, Li Y, et al. Effects of different swine manure to wheat straw ratios on antibiotic resistance genes and the microbial community structure during anaerobic digestion. Bioresour Technol. 2017;231:1–8. 10.1016/j.biortech.2017.01.054.Search in Google Scholar PubMed

[176] Howes SA. The effect of thermophilic anaerobic digestion on ceftiofur and antibiotic resistant gene concentrations in dairy manure. Va Tech. 2017. https://vtechworks.lib.vt.edu/handle/10919/86532. Accessed 20 October 2020.Search in Google Scholar

[177] Pu C, Liu H, Ding G, Sun Y, Yu X, Chen J, et al. Impact of direct application of biogas slurry and residue in fields: In situ analysis of antibiotic resistance genes from pig manure to fields. J Hazard Mater. 2018;344:441–9. 10.1016/j.jhazmat.2017.10.031.Search in Google Scholar PubMed

[178] Xu R, Yang Z-H, Wang Q-P, Bai Y, Liu J-B, Zheng Y, et al. Rapid startup of thermophilic anaerobic digester to remove tetracycline and sulfonamides resistance genes from sewage sludge. Sci Total Environ. 2018;612:788–98. 10.1016/j.scitotenv.2017.08.295.Search in Google Scholar PubMed

[179] Diehl DL, LaPara TM. Effect of temperature on the fate of genes encoding tetracycline resistance and the integrase of class 1 integrons within anaerobic and aerobic digesters treating municipal wastewater solids. Environ Sci Technol. 2010;44(23):9128–33. 10.1021/es102765a.Search in Google Scholar PubMed

[180] Ma Y, Wilson CA, Novak JT, Riffat R, Aynur S, Murthy S, et al. Effect of various sludge digestion conditions on sulfonamide, macrolide, and tetracycline resistance genes and class I integrons. Environ Sci Technol. 2011;45(18):7855–61. 10.1021/es200827t.Search in Google Scholar PubMed

[181] Sun W, Qian X, Gu J, Wang X-J, Duan M-L. Mechanism and effect of temperature on variations in antibiotic resistance genes during anaerobic digestion of dairy manure. Sci Rep. 2016;6(1):1–9. 10.1038/srep30237.Search in Google Scholar PubMed PubMed Central

[182] Huang H, Chen Y, Zheng X, Su Y, Wan R, Yang S. Distribution of tetracycline resistance genes in anaerobic treatment of waste sludge: The role of pH in regulating tetracycline resistant bacteria and horizontal gene transfer. Bioresour Technol. 2016;218:1284–89. 10.1016/j.biortech.2016.07.097.Search in Google Scholar PubMed

[183] Zhang L, Gu J, Wang X, Zhang R, Tuo X, Guo A, et al. Fate of antibiotic resistance genes and mobile genetic elements during anaerobic co-digestion of Chinese medicinal herbal residues and swine manure. Bioresour Technol. 2018;250:799–805. 10.1016/j.biortech.2017.10.100.Search in Google Scholar PubMed

[184] Wang R, Chen M, Feng F, Zhang J, Sui Q, Tong J, et al. Effects of chlortetracycline and copper on tetracyclines and copper resistance genes and microbial community during swine manure anaerobic digestion. Bioresour Technol. 2017;238:57–69. 10.1016/j.biortech.2017.03.134.Search in Google Scholar PubMed

[185] Zhang W, Huang MH, Qi FF, Sun PZ, Van Ginkel SW. Effect of trace tetracycline concentrations on the structure of a microbial community and the development of tetracycline resistance genes in sequencing batch reactors. Bioresour Technol. 2013;150:9–14. 10.1016/j.biortech.2013.09.081.Search in Google Scholar PubMed

[186] Huang X, Zheng J, Tian S, Liu C, Liu L, Wei L, et al. Higher temperatures do not always achieve better antibiotic resistance gene removal in anaerobic digestion of swine manure. Appl Environ Microbiol. 2019;85(7):e02878-18. 10.1128/aem.02878-18.Search in Google Scholar PubMed PubMed Central

[187] Lin Q, De Vrieze J, Li J, Li X. Temperature affects microbial abundance, activity and interactions in anaerobic digestion. Bioresour Technol. 2016;209:228–36. 10.1016/j.biortech.2016.02.132.Search in Google Scholar PubMed

[188] Ghosh S, Ramsden SJ, LaPara TM. The role of anaerobic digestion in controlling the release of tetracycline resistance genes and class 1 integrons from municipal wastewater treatment plants. Appl Env Microbiol. 2009;84(4):791–6. 10.1007/s00253-009-2125-2.Search in Google Scholar PubMed

[189] Sun W, Gu J, Wang X, Qian X, Peng HJ. Solid-state anaerobic digestion facilitates the removal of antibiotic resistance genes and mobile genetic elements from cattle manure. Bioresource Technol. 2018;274:287–95. 10.1016/j.biortech.2018.09.013.Search in Google Scholar PubMed

[190] Resende JA, Diniz C, Silva V, Otenio M, Bonnafous A, Arcuri P, et al. Dynamics of antibiotic resistance genes and presence of putative pathogens during ambient temperature anaerobic digestion. 2014;117(6):1689–99. 10.1111/jam.12653.Search in Google Scholar PubMed

[191] Hameed SA, Riffat R, Li B, Naz I, Badshah M, Ahmed S, et al. Microbial population dynamics in temperature‐phased anaerobic digestion of municipal wastewater sludge. J Chem Technol Biotechnol. 2019;94(6):1816–31. 10.1002/jctb.5955.Search in Google Scholar

[192] Wu Y, Cui E, Zuo Y, Cheng W, Chen H. Fate of antibiotic and metal resistance genes during two-phase anaerobic digestion of residue sludge revealed by metagenomic approach. Environ Sci Pollut Res. 2018;25(14):13956–63. 10.1007/s11356-018-1598-x.Search in Google Scholar PubMed

[193] Luo T, Wang Y, Pandey P. The removal of moisture and antibiotic resistance genes in dairy manure by microwave treatment. Environ Sci Pollut Res. 2020;28:1–9. 10.1007/s11356-020-10986-8.Search in Google Scholar PubMed

[194] Li HL, Wu CY, Tang AP, Tong J, Wei YS. Occurrence of antibiotic resistance genes and bacterial community structure of different sludge samples during microwave pretreatment-anaerobic digestion. Huan Jing Ke Xue Huanjing Kexue. 2021;42(1):323–32. 10.13227/j.hjkx.202006079.Search in Google Scholar PubMed

[195] Tian Z, Chi Y, Yu B, Yang M, Zhang Y. Thermophilic anaerobic digestion reduces ARGs in excess sludge even under high oxytetracycline concentrations. Chemosphere. 2019;222:305–13. 10.1016/j.chemosphere.2019.01.139.Search in Google Scholar PubMed

[196] Sun W, Gu J, Wang X, Qian X, Tuo X. Impacts of biochar on the environmental risk of antibiotic resistance genes and mobile genetic elements during anaerobic digestion of cattle farm wastewater. Bioresour Technol. 2018;256:342–49. 10.1016/j.biortech.2018.02.052.Search in Google Scholar PubMed

[197] Zhang Y, Yang Z, Xiang Y, Xu R, Zheng Y, Lu Y, et al. Evolutions of antibiotic resistance genes (ARGs), class 1 integron-integrase (intI1) and potential hosts of ARGs during sludge anaerobic digestion with the iron nanoparticles addition. Sci Total Environ. 2020;724:138248. 0.1016/j.scitotenv.2020.138248.Search in Google Scholar

[198] Zhang R, Gu J, Wang X, Li Y, Liu J, Lu C, et al. Response of antibiotic resistance genes abundance by graphene oxide during the anaerobic digestion of swine manure with copper pollution. Sci Total Environ. 2019;654:292–9. 10.1016/j.scitotenv.2018.11.094.Search in Google Scholar

[199] Shi L, Hu Z, Simplicio WS, Qiu S, Xiao L, Harhen B, et al. Antibiotics in nutrient recovery from pig manure via electrodialysis reversal: Sorption and migration associated with membrane fouling. J Membr Sci. 2020;597:117633. 10.1016/j.memsci.2019.117633.Search in Google Scholar

[200] Li S, Liu Y, Ge R, Yang S, Zhai Y, Hua T, et al. Microbial electro-Fenton: A promising system for antibiotics resistance genes degradation and energy generation. Sci Total Environ. 2020;699:134160. 10.1016/j.scitotenv.2019.134160.Search in Google Scholar

[201] Ngigi AN, Ok YS, Thiele-Bruhn S. Biochar affects the dissipation of antibiotics and abundance of antibiotic resistance genes in pig manure. Bioresour Technol. 2020;315:123782. 10.1016/j.biortech.2020.123782.Search in Google Scholar

[202] Quejigo JR, Tejedor-Sanz S, Schroll R, Esteve-Núñez A. Electrodes boost microbial metabolism to mineralize antibiotics in manure. Bioelectrochemistry. 2019;128:283–90. 10.1016/j.bioelechem.2019.04.008.Search in Google Scholar

[203] Li H, Li B, Ma J, Ye J, Guo P, Li L. Fate of antibiotic-resistant bacteria and antibiotic resistance genes in the electrokinetic treatment of antibiotic-polluted soil. Chem Eng J. 2018;337:584–94. 10.1016/j.cej.2017.12.154.Search in Google Scholar

[204] Ma J, Gu J, Wang X, Peng H, Wang Q, Zhang R, et al. Effects of nano-zerovalent iron on antibiotic resistance genes during the anaerobic digestion of cattle manure. Bioresour Technol. 2019;289:121688. 10.1016/j.biortech.2019.121688.Search in Google Scholar

[205] Staley ZR, Tuan CY, Eskridge KM, Li X. Using the heat generated from electrically conductive concrete slabs to reduce antibiotic resistance in beef cattle manure. Sci Total Environ. 2021;768:144220. 10.1016/j.scitotenv.2020.144220.Search in Google Scholar

[206] Sawant AA, Sordillo LM, Jayarao BM. A survey on antibiotic usage in dairy herds in Pennsylvania. J Dairy Sci. 2005;88(8):2991–9. 10.3168/jds.S0022-0302(05)72979-9.Search in Google Scholar

[207] OIE. OIE list of antimicrobial agents of veterinary importance. World Organisation for Animal Health; 2015. https://www.oie.int/fileadmin/Home/eng/Our_scientific_expertise/docs/pdf/AMR/A_OIE_List_antimicrobials_May2018.pdf. Accessed 20 October 2020.Search in Google Scholar

[208] Garneau-Tsodikova S, Labby KJ. Mechanisms of resistance to aminoglycoside antibiotics: Overview and perspectives. Medchemcomm. 2016;7(1):11–27. 10.1039/C5MD00344J.Search in Google Scholar PubMed PubMed Central

[209] Li X-Z, Plésiat P, Nikaido H. The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev. 2015;28(2):337–418. 10.1128/CMR.00117-14.Search in Google Scholar

[210] Ramirez MS, Nikolaidis N, Tolmasky M. Rise and dissemination of aminoglycoside resistance: the aac (6′)-Ib paradigm. Front Microbiol. 2013;4:121. 10.3389/fmicb.2013.00121.Search in Google Scholar

[211] Wachino J, Arakawa Y. Exogenously acquired 16S rRNA methyltransferases found in aminoglycoside-resistant pathogenic Gram-negative bacteria: An update. Drug Resist Updat. 2012;15(3):133–48. 10.1016/j.drup.2012.05.001.Search in Google Scholar

[212] Bradford PA. Extended-spectrum β-lactamases in the 21st century: characterization, epidemiology, and detection of this important resistance threat. Clin Microbiol Rev. 2001;14(4):933–51. 10.1128/CMR.14.4.933-951.2001.Search in Google Scholar

[213] Walsh TR. Emerging carbapenemases: A global perspective. Int J Antimicrob Agents. 2010;36:S8–S14. 10.1016/S0924-8579(10)70004-2.Search in Google Scholar

[214] Marsik FJ, Nambiar S. Review of carbapenemases and AmpC-beta lactamases. Pediatric Infect Dis J. 2011;30(12):1094–5. 10.1097/INF.0b013e31823c0e47.Search in Google Scholar PubMed

[215] Rice LB. Mechanisms of resistance and clinical relevance of resistance to β-lactams, glycopeptides, and fluoroquinolones. Mayo Clin Proc. 2012;87(2):198–208. 10.1016/j.mayocp.2011.12.003.Search in Google Scholar PubMed PubMed Central

[216] Schwarz S, Kehrenberg C, Doublet B, Cloeckaert A. Molecular basis of bacterial resistance to chloramphenicol and florfenicol. FEMS Microbiol Rev. 2004;28(5):519–42. 10.1016/j.femsre.2004.04.001.Search in Google Scholar PubMed

[217] Roberts MC. Update on macrolide–lincosamide–streptogramin, ketolide, and oxazolidinone resistance genes. FEMS Microbiol Lett. 2008;282(2):147–59. 10.1111/j.1574-6968.2008.01145.x.Search in Google Scholar PubMed

[218] Mitsuyama J, Itoh Y, Takahata M, Okamoto S, Yasuda T. In vitro antibacterial activities of tosufloxacin against and uptake of tosufloxacin by outer membrane mutants of Escherichia coli, Proteus mirabilis, and Salmonella typhimurium. Antimicrob Agents Chemother. 1992;36(9):2030–6. 10.1128/aac.36.9.2030.Search in Google Scholar

[219] Jacoby GA. Mechanisms of resistance to Quinolones. Clin Infect Dis. 2005;41(Supplement_2):S120–S26. 10.1086/428052.Search in Google Scholar PubMed

[220] Ruiz J. Mechanisms of resistance to quinolones: target alterations, decreased accumulation and DNA gyrase protection. J Antimicrob Chemother. 2003;51(5):1109–17. 10.1093/jac/dkg222.Search in Google Scholar PubMed

[221] Sköld O. Sulfonamide resistance: mechanisms and trends. Drug Resistance Updates. 2000;3(3):155–60. 10.1054/drup.2000.0146.Search in Google Scholar PubMed

[222] Roberts MC. Update on acquired tetracycline resistance genes. FEMS Microbiol Lett. 2005;245(2):195–203. 10.1016/j.femsle.2005.02.034.Search in Google Scholar PubMed

[223] Sharma R, Larney FJ, Chen J, Yanke LJ, Morrison M, Topp E, et al. Selected antimicrobial resistance during composting of manure from cattle administered sub-therapeutic antimicrobials. J Environ Qual. 2009;38(2):567–75. 10.2134/jeq2007.0638.Search in Google Scholar PubMed

[224] Yu Z, Michel FC, Hansen G, Wittum T, Morrison MJA. Development and application of real-time PCR assays for quantification of genes encoding tetracycline resistance. Appl Environ Microbiol. 2005;71(11):6926–33. 10.1128/AEM.71.11.6926-6933.2005.Search in Google Scholar PubMed PubMed Central

[225] Zhang Y, Yang Z, Xiang Y, Xu R, Zheng Y, Lu Y, et al. Evolutions of antibiotic resistance genes (ARGs), class 1 integron-integrase (intI1) and potential hosts of ARGs during sludge anaerobic digestion with the iron nanoparticles addition. Sci Total Environ. 2020 Jul 1;724:138248.10.1016/j.scitotenv.2020.138248Search in Google Scholar PubMed

[226] Wang Q, Gu J, Wang X, Ma J, Hu T, Peng H, et al. Effects of nano-zerovalent iron on antibiotic resistance genes and mobile genetic elements during swine manure composting. Environ Pollut. 2020 Mar 1;258:113654.10.1016/j.envpol.2019.113654Search in Google Scholar PubMed

[227] Amarasiri M, Sano D, Suzuki S. Understanding human health risks caused by antibiotic resistant bacteria (ARB) and antibiotic resistance genes (ARG) in water environments: Current knowledge and questions to be answered. Crit Rev Environ Sci Technol. 2020 Oct 1;50(19):2016–59.10.1080/10643389.2019.1692611Search in Google Scholar

[228] Ecotox Knowledge. ECOTOX | Home (epa.gov); 2022. (accessed on 7/24/2022).Search in Google Scholar

[229] Zhang AN, Gaston JM, Dai CL, Zhao S, Poyet M, Groussin M, et al. An omics-based framework for assessing the health risk of antimicrobial resistance genes. Nat Commun. 2021 Aug 6;12(1):1.10.1038/s41467-021-25096-3Search in Google Scholar PubMed PubMed Central

[230] Kim M, Kienast Y, Hatt JK, Kirby AE, Konstantinidis KT. Metagenomics indicate that public health risk may be higher from flooding following dry versus rainy periods. Environ Microbiol Rep. 2022 Apr;14(2):265–73.10.1111/1758-2229.13047Search in Google Scholar PubMed

[231] Cadena M, Durso LM, Miller DN, Waldrip HM, Castleberry BL, Drijber RA, et al. Tetracycline and sulfonamide antibiotic resistance genes in soils from Nebraska organic farming operations. Front Microbiol. 2018 Jun 28;9:1283.10.3389/fmicb.2018.01283Search in Google Scholar PubMed PubMed Central

Received: 2021-09-07
Revised: 2022-07-25
Accepted: 2022-08-06
Published Online: 2022-09-15

© 2022 Yi Wang et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 21.5.2024 from https://www.degruyter.com/document/doi/10.1515/opag-2022-0129/html
Scroll to top button