1,719
Views
306
CrossRef citations to date
0
Altmetric
Review

Horizontal gene transfer in the human gastrointestinal tract: potential spread of antibiotic resistance genes

Pages 167-176 | Published online: 20 Jun 2014

Abstract

Bacterial infections are becoming increasingly difficult to treat due to widespread antibiotic resistance among pathogens. This review aims to give an overview of the major horizontal transfer mechanisms and their evolution and then demonstrate the human lower gastrointestinal tract as an environment in which horizontal gene transfer of resistance determinants occurs. Finally, implications for antibiotic usage and the development of resistant infections and persistence of antibiotic resistance genes in populations as a result of horizontal gene transfer in the large intestine will be discussed.

Introduction

The emergence of antibiotic resistant infections is considered to be a major global health issue.Citation1 It is feared that the golden age of antibiotic therapy is coming to an end, and that we will soon return to a pre-antibiotic era, where there are limited successful treatments for infectious diseases caused by bacteria. These resistant infections are costly to society in economic impact as well as in the morbidity and mortality of affected individuals. Taking into account just methicillin-resistant Staphylococcus aureus and third-generation cephalosporin-resistant Escherichia coli infections alone, each year there are thousands of deaths, exceedingly more infections, and millions of international dollars spent on health care costs.Citation2 Even though infectious diseases still remain a leading cause of death worldwide,Citation3 antibiotic development by pharmaceutical companies has decreased dramatically, only contributing to about 0.2% of new drug development.Citation4 Evolution of antibiotic resistance continues, especially that of multidrug resistant strains, complicating treatment options.Citation1

Bacterial populations susceptible to antibiotics become resistant either through genetic mutation or through horizontal transfer and expression of resistance genes from other strains, either distantly or closely related. The human gastrointestinal tract provides an ideal combination of factors for antibiotic resistance genes to arise and spread through bacterial populations. One of these factors is high cell density. Other factors favoring spread of resistant infections include antibiotic exposure and subsequent selection followed by the innate ability for gene transfer through a variety of different mechanisms.

The aim of this review is to provide a brief overview of the bacterial processes that facilitate horizontal gene transfer, and to propose how this phenomenon may be taking place in the lower human gastrointestinal tract, specifically the large intestine, intensifying the antibiotic resistance crisis we are currently facing and setting the stage for future resistant infections.

Mechanisms and evolution of horizontal gene transfer

The three major mechanisms by which bacteria transfer genes horizontally are conjugation, natural transformation, and transduction. Conjugation is the most studied mechanism of horizontal gene transfer in the human intestine or otherwise.Citation5Citation7 Conjugation is the transfer of DNA fragments that can be very small, up to large chromosomes. The general order of the events of conjugation is a) cell-to-cell contact, b) mating pair formation, and finally c) transfer of plasmid DNA through a conjugative pilus. The conjugative pilus is considered to be machinery of the Type IV secretion system.Citation8 The genes for conjugative machinery are encoded by autonomously replicating plasmids or by integrative conjugative elements in the chromosome.Citation9

Conjugation requires cell-to-cell contact, usually via a pilus or pore that forms a channel that allows for the passage of plasmids. Generally, plasmids are covalently closed, circular molecules of DNA that replicate independently from the host chromosome. Some plasmids are conjugative, that is self-transmissible, while others are not. Conjugative plasmids harbor genes for the transfer machinery. These non-conjugative plasmids are mobilizable if they can be transferred through conjugation when a helper self-transmissible plasmid is present.Citation10 An in-depth model for the evolution of the mechanisms of conjugation has recently been elucidated, and the reader is referred to Guglielmini et al for further details.Citation11

The role of conjugation in horizontal gene transfer is further complicated by the fact that other mobile genetic elements besides plasmids can be transferred by conjugative machinery. Conjugative transposons are transposons which excise themselves from covalently closed circular DNA, such as a plasmid or chromosome, and integrate into another location in the same genome or are transferred by conjugation and then integrated into a different genome. These elements can also mobilize non conjugative plasmids or other DNA sequences in a cell and transfer them.Citation12 Another level of complexity is added when other mobile genetic elements such as integrons or integrating conjugative elements (ICE) are considered in tandem with plasmids and conjugative transposons.Citation13 Integrons are mobile genetic elements that encode integrase and are capable of site-specific recombination and typically carry antibiotic resistance genes.Citation14 ICE are transferred through cell-to-cell contact and can integrate into the chromosome of the recipient. SXT, an approximately 100 kb fragment of DNA, is one of the most widely studied of these elements and confers resistance to chloramphenicol, sulfamethoxazole, and trimethoprim and has both plasmid and phage sequences.Citation9,Citation15 Virtually any gene in intestinal bacteria can acquire the ability to be mobilized and transferred via a mobile genetic element.Citation16

The main mechanism of horizontal transfer of plasmids is through conjugation.Citation17 However, natural transformation does allow for the uptake of plasmid DNA as well as chromosomal DNA. Because of this, it is widely accepted that natural transformation is a major mechanism in the evolution of microbes. However, it is still unclear as to how and why the process itself evolved. Natural transformation is the process whereby bacterial cells take up free DNA from the environment and incorporate it into their genomes. There are over 80 known different species of naturally transformable bacteria, which includes members from different lineages including photolithotrophs, chemolithotrophs, heterotrophs, and methylotrophs.Citation18,Citation19 At least a dozen of these strains are pathogens which have been isolated from the clinical setting.Citation18 When evaluating bacterial or archaeal strains as naturally transformable, laboratory conditions known to induce competence are used. There are unquestionably many more strains that are naturally transformable, but have not been designated as such, because it is still unknown what environmental factor or combinations of factors trigger competence development. The evolutionary origin of natural transformation is unclear because the process is widely distributed over the major clades of bacteria and yet members of the same genus or even species can differ in their ability to be naturally transformed. This suggests that either there is a common origin for transformation that has been lost many times or that it has arisen independently on multiple occasions.Citation20

The overall process of natural genetic transformation can be broken down into six major steps.Citation18 They are a) DNA is released from donor cells, b) DNA is dispersed, c) DNA persists in the environment, d) recipient cells become competent for DNA uptake, e) competent cells interact with and take up the DNA, incorporating it into the genome either through homologous or illegitimate recombination, and finally f) genes encoded on the donor DNA are expressed in the recipient cells.Citation18 In the former three steps, the donor cell need not be alive or even in close proximity to the recipient, whereas the recipient cell is actively involved in the latter three steps.

There are five models explaining reasons why bacteria take up DNA from the environment. With the exception of Neisseria gonorrhea,Citation21 all known transformable bacteria must be induced to competence. Competence is the physiological state that allows for uptake of DNA into a bacterial cell. During competence development, genes for protein components of the uptake machinery are expressed. There are many triggers for competence development, which also makes it difficult to understand the evolutionary origins of the process.

The first model suggests that DNA is taken up into the bacterial cell to be used as a nutrient source.Citation22 This model is supported by studies that show some bacterial strains become competent to take up DNA under nutrient-limited conditions.Citation23 Many bacterial species, such as Gallibacterium anatis,Citation24 Haemophilus influenza,Citation25 and Aeromonas salmonicidaCitation26 are induced to competence in vitro by transferring the cultures from a rich medium to a starvation medium. More specifically, H. influenzae becomes competent as a result of purine starvation.Citation27

The second model explaining the evolutionary origins of competence development and natural transformation is that natural transformation evolved so that transformed DNA molecules could serve as templates for DNA repair. Bacillus subtilis cells exposed to ultraviolet light have greater survivorship when they are treated with DNA after damage than if they are treated with DNA before damage. The added DNA presumably serves as a template in recombinational repair.Citation28 Competence, specifically the com regulon, can be induced in Streptococcus pneumoniae upon exposure to mitomycin C, a DNA-damaging agent.Citation29

A third model is that natural transformation evolved to allow the cell to purposely obtain novel genetic information to increase genetic diversity in order to respond more favor-ably to natural selection.Citation30 However, a study on transform-ability in Acinetobacter baylyi revealed that both competent strains and noncompetent strains adapted to laboratory conditions at the same rate.Citation31 Competence did not confer any discernible advantage, and the competent lineages even evolved a lower level of transformability under laboratory conditions. Engelstädter and Moradigaravand recently proposed a complementary model in which competence evolved because it allows bacterial cells to take up DNA from the environment that has persisted for an indefinite period of time to incorporate genes into genomes in order that the cell may revert to a prior genetic state.Citation32

The fourth model is that natural transformation evolved and is maintained because of episodic stressful conditions that may indirectly select for non-growing competent cells.Citation33 Competent populations of cells, like Bacillus subtilis, do not grow for several hours.Citation34 This is considered to be a “persister” state. The persister state was first recognized in 1944 in surviving Staphylococcus cells that were treated with penicillin. These cells survived because they were not growing and unaffected by the antibiotic, not because they were mutants with altered antibiotic targets.Citation35 These cells are now considered to be tolerant to antibiotics instead of resistant to them.Citation36 Episodic selection of persister cells occurs when the cell population experiences stressful conditions that kill the growing cells at a higher rate than the non-growing persister cells. If these persister cells are also competent as some are, then natural transformation ability is also indirectly selected by the stress.Citation33 This hypothesis has been supported using antibiotics (penicillin-G) as the episodic selection in populations of B. subtilis where the competent cells had better survivability than the non-competent mutants.Citation33

The fifth, and final, model posits that natural transformation is an unintended consequence of the type IV pili actions of twitching motility and cell adhesion.Citation37 Type IV pili are known to take up free DNA during natural transformation.Citation38,Citation39 As the cells move across surfaces using their type IV pili for twitching motility, it is possible that they accidentally bind DNA at PilC, the tip of the pilus. As the pilus retracts, the bound DNA blocks the PilQ pore present in the outer membrane and passes through it after one strand is degraded by nucleases. In this model, environmental stress triggers the SOS response (an error-prone DNA repair system), inducing the expression of type IV pili so that the cell can move using twitching motility to a more favorable environment. Free DNA is bound and taken up by the cell as a consequence of the active type IV pili.Citation37 In this model, natural transformation ability is accidental, although it can be beneficial when the cell is able to use the new DNA as a nutrient source or as a template for genome repair.Citation37 Whatever the biological reason for the uptake, one consequence of transformation is a changed genotype of the recipient cell. Transformation is widespread among microorganisms and has a lasting impact on bacterial genomes that is difficult, if not impossible, to assess.Citation40

Transduction is another mechanism of horizontal gene transfer that has shaped the evolution of bacterial genomes. The abundance of information about antibiotic resistance gene transfer via bacteriophages is limited when compared to natural transformation and conjugation. However, the role of bacteriophages in transducing antibiotic resistance genes is now becoming clearer. Bacteriophages are viruses that infect bacteria and are considered to be temperate if they integrate into the host genome and become prophages until environmental conditions trigger their lytic growth.Citation41 This state can last indefinitely and can confer resistance to infection by bacteriophages of the same group.Citation41

Transduction occurs when a bacteriophage that has previously replicated in another bacterial cell packages a portion of the host genome (donor) into the phage head and transfers the genes to another (recipient) bacterial cell. Transduction is thought to be an unevolved process that occurs as a consequence of mistakes in excision of bacteriophage DNA from the donor genome. Transduction can be classified as either specialized or generalized. Specialized transduction occurs when only bacterial DNA adjacent to the attachment site of a temperate bacteriophage, as in λ in E. coli, is accidentally packaged into the bacteriophage and subsequently transferred to a recipient cell. Generalized transduction occurs when any gene from a host is randomly packaged along with viral DNA into a bacteriophage head, such as occurs in P22 of Salmonella, and then transferred to a new recipient. The donor strain and the recipient strain need not be in close proximity in either geographical location or time to one another, as bacteriophages can persist in the environment for varying time periods.Citation42 Virtually any DNA sequence, including antibiotic resistance, found in a bacterial genome can be transferred.Citation43 This includes chromosomal sequences and mobile genetic elements like plasmids, transposons, and insertion elements.Citation44

Transduction as a significant mechanism of horizontal gene transfer in naturally occurring environments has traditionally been underestimated. A metagenomic analysis of viromes suggests that functional bacterial genes of all types exist in up to 50% to 60% of bacteriophages and that these particles can serve as a reservoir for genes in a diversity of environments and also a mechanism for their transfer among bacteria.Citation45 Transduction as an important mechanism of horizontal gene transfer has gained support with significant studies showing that antibiotic resistance genes are found in abundance in phage particles in nature.Citation42 Transduction has only recently been considered as playing a significant role in the transfer of antibiotic resistance among potential pathogens as demonstrated from Enterococcus gallinarum to Enterococcus faecalis during in vitro experiments.Citation46 Other in vitro experiments with Salmonella transducing bacteriophages demonstrated that ampicillin, chloramphenicol, and tetracycline resistance genes could be transduced.Citation43

Role of horizontal gene transfer in the spread of antibiotic resistance genes in the large intestine

Conjugation, natural transformation, and transduction have served as important mechanisms in the evolution of bacteria. However, there is evidence that these mechanisms play a role in evolution that is currently occurring in the large intestine under the selective pressure of antibiotics. The most common mechanism of horizontal gene transfer in other natural environments is thought to be by plasmids through conjugation.Citation17 It stands to reason that this is also true for the gastrointestinal tract.

The lower gastrointestinal tract is the largest microbial community in the human host, consisting of up to 1014 individual bacterial cells of over 500 species from nine bacterial divisions, including Actinobacteria, Bacteroidetes, Cyanobacteria, Firmicutes, Fusobacteria, Proteobacteria, Spirochaeates, Verrucomicrobia, and VadinBE97.Citation47,Citation48,Citation49 Biofilms are found in the human intestinal tract and are thought to be ideal environments for horizontal gene transfer because they allow for high bacterial density and physical protection for the cells. The biofilm environment supports the cell-to-cell contact needed for conjugation to occur.Citation50,Citation51 E. coli cells are found as coccoid cells embedded in the mucosal layer of the large intestine.Citation52,Citation53 As a side note, populations of Streptococcus pyogenes growing in laboratory biofilms become naturally transformable.Citation54 These biofilms consist of many species and have a greater resistance to antibiotics than their free-living counterparts.Citation55,Citation56

Salyers et alCitation57 proposed the resistance gene reservoir hypothesis in which bacteria of the human gastrointestinal tract serve as a reservoir for antibiotic resistance genes that can be transferred to other resident intestinal bacteria or transient bacteria that pass through, but do not colonize, the large intestine for a significant period of time. Several recent studies have demonstrated the magnitude of antibiotic resistance gene reservoir in the human intestinal tract. The most current version of the Antibiotic Resistance Genes Database (ARDB) lists over 23,000 known gene and protein sequences for at least 240 different antibiotics (version 1.1).Citation58 A study comparing the gut metagenomes from 275 individuals to the sequences deposited in the ARDB database showed resistance to 53 antibiotics. The study also found multiple antibiotic resistance genes on contigs with integrase and transposase, indicating the possibility that these clusters are part of mobile genetic elements, such as conjugative transposons. Furthermore, 97% of the gut metagenomes harbored resistance genes to tetracycline, 95% to bacitracin, and 95% to tetracycline.Citation59 Another metagenome analysis evaluated the gut microbiome from 162 individuals (encompassing 4.1 million genes) and found 1,093 antibiotic resistance genes.Citation60 A single erythromycin gene type, ermB, was found in all but one of the samples. A gene, b12e_cfxa, conferring resistance to cephalosporin and bacA (recently renamed as uppP), conferring resistance to bacitracin, and vanRG, conferring resistance to vancomycin, were also found at high relative abundance in the samples.Citation61 The human intestinal tract is a reservoir for more antibiotic resistance genes than any other studied natural environment, such as soil, marine, or lake environments.Citation61,Citation60

Forslund et al compared metagenomes analysis from 252 fecal samples from individuals from Spain, Denmark, and the United States.Citation62 The antibiotic resistance genes were more abundant in all metagenomes for those antibiotics that have been used in the clinical setting longer and for those that are approved by the national governments for use in livestock than the antibiotics introduced later and not used in animals. In fact, resistance genes for antibiotics approved for use in livestock are the most abundant resistance genes in the human gut microbiome.Citation62 This evidence supports the “farm-to-fork” hypothesis that the non-therapeutic use of antibiotics in animal husbandry leads to increased antibiotic resistance in humans through consumption of affected food or water.Citation63,Citation64 Antibiotics can pass into the human gastrointestinal tract through the consumption of undercooked or precooked meat exposing the resident microflora and the transient bacterial cells to the effects of the antibiotics.

Specifically, a source of antibiotic resistance genes in the human large intestine is the lactic acid bacteria. These bacteria convert fermentable carbohydrates into lactic acid. They are typically Gram-positive, catalase-negative, have low C + G content, and include Enterococcus, Lactobacillus, Lactococcus, Leuconostoc, Pediococcus, and Streptococcus.Citation65 They are used in the production of dairy products such as yogurt, buttermilk, and some cheeses as well as in the production of sausage, hams, and wine.Citation65 Lactic acid bacteria colonize the human large intestine but can cause opportunistic infections in immunocompromised patients.Citation65 The US Food and Drug Administration (FDA) declared lactic acid bacteria as “Generally Regarded As Safe” (GRAS) in terms of ingestion of these microbes, but there are new questions as to whether these strains may serve as a source of antibiotic resistance genes. Atypical isolates resistant to erythromycin and tetracycline have been foundCitation66, as well as strains harboring plasmids encoding resistance to chloramphenicol that were derived from staphylococcal sequences and a tetracycline gene that is identical to one found in Listeria monocytogenes.Citation67

Further in vivo and in vitro studies into transmissibility of genes among lactobacilli have demonstrated that, in the absence of selection, tetracycline resistance is not widely transferred.Citation68,Citation69 However, in an in vivo study, an Enterococcus faecium isolate from animal origin transferred vanA, a gene for vancomycin-resistance, to a vancomycin-susceptible E. faecium of human origin in the intestines of half (3/6) of the human volunteers.Citation70 The donor strain from animal origin did not persist and disappeared within two weeks of inoculation. This lends further support to the “farm-to-fork” hypothesis in which transient strains can be ingested in food and then transfer antibiotic resistance to commensals in the human intestine.Citation70

Another similar in vivo study where human volunteers ingested E. coli shows similar results. In this case, instead of the transfer of genes between the two administered strains in the gastrointestinal tract as the experiments were designed, sulfonamide and ampicillin resistances were transferred on a conjugative plasmid from the indigenous E. coli to the administered E. coli.Citation71

Reports where horizontal gene transfer was demonstrated to have occurred in the lower gastrointestinal tract are becoming more frequent. An analysis of a nosocomial outbreak of Enterobacter cloacae revealed that there was probable conjugal transfer of an OXA-48 (carbapenem-resistance) encoding plasmid from E. cloacae to other members of Enterobacteriaceae in the intestines of a patient, which was then spread to other patients.Citation72 Another report showed that an E. coli donor strain transferred ampicillin resistance on a plasmid to another E. coli strain in the intestines of an infant. It is presumed that treatment with ampicillin for a urinary tract infection provided the selection needed in order for the donor density to increase and favor the transfer of the plasmid to the recipient strain.Citation73

Another case study reported the presumable transfer of a multidrug resistance plasmid from Klebsiella pneumoniae to E. coli in the gastrointestinal tract of a patient. Before treatment with ertapenem, metronidazole, colistin, and vancomycin, no carbapenem-resistant E. coli could be isolated from the patient, but after treatment a resistant strain was found. This resistant E. coli had the Klebsiella pneumoniae plasmid, blaKPC-3, which conferred resistance to cephalosporins, monobactams, and carbapenems. Again, antibiotic treatment is thought to have triggered the transfer and maintained the selection of the resistance plasmid.Citation74

Other studies have demonstrated the conjugal transfer of resistance plasmids in the gastrointestinal tracts of animal models. A plasmid encoding resistance to β-lactams, aminoglycosides, tetracyclines, sulfonamides, and phenicols was transferred from Serratia liquefaciens to E. coli in the intestinal tracts of gnotobiotic mice with human fecal flora.Citation75 Gentamicin resistance was transferred from Enterococcus faecalis to E. coli,Citation76 while aminoglycoside and macrolide resistance was transferred via conjugation among Enterococcus strains.Citation77

The conjugation of plasmids encoding antibiotic resistance machinery has led to worldwide dissemination of these genes, particularly in Enterobacteriaceae. These plasmids can encode resistance to β-lactamases (CMY, DHA, GES, LAP, NDM, SHV, TEM), extended spectrum β-lactamases (CTX, VEB), metallo β-lactamases (IMP), carbapenemases (KPC, VIM); quinolone resistance (Qep, Qnr), aminoglycoside resistance (AAC, Arm, RmtB), tetracycline resistance (Tet), sulfonamide resistance (Sul), colistin resistance. They also encode the Cfr rRNA methyltransferase which confers resistance to phenicols, lincosamides, oxazolidinones, pleuromutilins, and streptogramin A.Citation78Citation82

A study evaluating fecal fosmid libraries for the presence of tetracycline resistance gene found a single individual to have distantly related bacteria harboring identical resistance sequences. The authors propose that this individual most likely acquired the genes through horizontal gene transfer in the gastrointestinal tract.Citation83

An investigation by Shoemaker et al revealed that Bacteroides strains have undergone extensive horizontal gene transfer over the past 30 years leading to widespread resistance to tetracycline and erythromycin encoded by just a few identical determinants carried on conjugal elements.Citation84 It is hypothesized that the human colon provides an environment in which Bacteroides can engage in horizontal gene transfer within the genus and across distantly related genera.Citation84

Horizontal gene transfer in the large intestine occurs at levels that cannot be detected because of the complex nature of the microbial community structure both in diversity and in cell numbers. The physical and chemical conditions in this environment allow for natural transformation, conjugation, and transduction to occur. However, horizontal gene transfer through natural transformation may occur at greater frequencies than previously expected due to the presence of triggers of competence that we have not yet discovered. The importance of natural transformation in this environment, and possibly all environments, is underestimated.

Implications for antibiotic usage and the development of resistant infections

It has long been known that antibiotic resistance arises as a consequence of exposure to antibiotics. The resistant cells in a population have an advantage over sensitive cells when exposed to this strong selection. Therefore, the population becomes resistant to antibiotics. However, it is now known that antibiotics not only select for resistant populations through the clonal expansion of already-resistant cells, but they also create them by inducing the horizontal transfer of resistance genes.

Antibiotic exposure is a source of stress that can create stress-induced resistance among bacteria as part of the SOS response. The SOS response is an inducible DNA repair network, first discovered in E. coli by Radman in 1975.Citation85 Under the response, mutations and genetic exchanges within the cell increase in order for damaged DNA to be repaired and the cell to survive. As a consequence of these genetic manipulations, any gene sequence, including those for antibiotic targets can be altered, providing the possibility for evolution of resistance.Citation86 Furthermore, antibiotics that cause double-stranded breaks, such as ciprofloxacin induce the SOS response. The SOS response is both induced by antibiotics and allows for antibiotic resistance to arise.Citation86 Chromosomal mutations accumulate rapidly which, as a consequence, confer resistance, as demonstrated by Streptococcus uberis and Pseudomonas aeruginosa gaining resistance to multiple antibiotics when exposed to sub-lethal concentrations of single antibiotics.Citation87,Citation88 In a study with Acinetobacter oleivorans, it was demonstrated that treatment with norfloxacin provided selection of resistant variants, induced genetic changes which resulted in a beneficial mutation in the molecular target of norfloxacin, gyrA, and also caused an upregulation of the SOS response.Citation89

It is well established that antibiotic stress induces the SOS response, but for some of those bacterial strains that lack the response another mechanism is induced. Legionella pneumophila, a pathogen of human alveolar macrophages which lacks the SOS response, is induced to competence for natural genetic transformation by fluoroquinolones and other DNA-damaging antibiotics and chemicals.Citation90 The same is true for the human pathogen S. pneumoniae, which uses competence as a general response to stress, and has been shown to become competent under exposure to aminoglycoside and fluoroquinolone antibiotics.Citation29 Antibiotics can provide enough stress to induce competence in strains without the SOS response, and the environment of the gastrointestinal system is conducive to natural transformation. Presumably, DNA is present from recently lysed cells and could be protected from degradation by biofilms. Extracellular DNA is also a major constituent of most biofilms.Citation91 At this time it is still unknown if competence can be induced by antibiotics in other pathogenic strains that lack the SOS response, like Neisseria gonorrhoeae, Neisseria meningitides, and Campy-lobacter jejuni.Citation92

When the SOS response is triggered, it directly induces the activation of prophages to lytic bacteriophages, which has consequences on both evolution and the ecology of the microflora of the gastrointestinal tract.Citation93 For example, Burkholderia thailandensis (a soil microbe), in response to ciprofloxacin exposure, transcribed genes associated with the SOS response as well as those of a viable bacteriophage.Citation94

A study analyzing the intestinal microflora of mice showed that antibiotic administration of ciprofloxacin and ampicillin creates stressful conditions for bacteria, inducing the SOS response, which increases the abundance of antibiotic resistance genes found within the phage particles.Citation95 These phages can act as a reservoir of all types of genes in order to maintain the functional integrity of the intestinal microflora. Subsequent bacteriophage infection allows for the transduction of resistance genes. There is evidence that the favored life cycle of bacteriophages is lysogeny in the gastrointestinal tract.Citation55 However, in the “community shuffling” model, the SOS response causes induction of prophages to bacteriophages resulting in the lysing of bacteria, which in turn causes dysbiosis.Citation89,Citation55 Dysbiosis is a disturbance in the normal intestinal microflora and a precursor to chronic diseases, such as Crohn’s disease, ulcerative colitis, and irritable bowel syndrome.Citation96,Citation97 Higher abundance of bacteriophages has been found in gut wash samples of individuals with Crohn’s disease compared to those without disease.Citation98 This suggests that induction by antibiotics may assist in the development of acute infections as well as in chronic diseases, but more research must be done in order to test this hypothesis.

Induction of the SOS response has other implications for the development of resistant infections as well. The response can also induce the transfer of mobile genetic elements, such as has been noted for SXT, an integrating conjugating element, in Vibrio cholerae allowing for the transfer of the multidrug resistance genes.Citation99 Furthermore, in a study with Staphylococcus aureus, antibiotics induced the SOS response, which in turn activated the transduction of virulence genes.Citation100 This suggests that infection with transducing phages leads to more virulent pathogenic bacteria that are resistant to antibiotics.

It has long been believed that antibiotic-resistant bacterial cells have reduced fitness compared to their susceptible counterparts and that, in the absence of antibiotic selection, populations of antibiotic resistant strains will decrease, but, unfortunately, there is no solid evidence for this.Citation101 It is now known that metabolic cost of antibiotic resistance is not as high as once believed and that cells, through continued selection and evolution, are able to mitigate that cost.Citation101 Some cells do revert back to their previously susceptible state, but most cells will evolve compensatory mutations.Citation101 For example, a study with Salmonella typhimurium and nalidixic acid resistance, showed a compensatory mutation in gyrA that improves the fitness of the resistant strain.Citation102 Not all antibiotic-resistance genes carry with them an associated fitness cost. Another study with S. typhimurium, demonstrated that about half of their fluoroquinolone-resistant isolates had a selective advantage over the sensitive parent strain in the absence of the antibiotic, indicating that this type of resistance is not associated with a reduction in fitness.Citation103

An in vivo study in which Bacteroides strains were analyzed from fecal samples from human volunteers exposed to clindamycin showed that within 7 days they had acquired erm genes (for erythromycin resistance) through conjugation and were resistant to clindamycin. This pattern continued through at least 18 months after the initial antibiotic administration suggesting that any fitness cost was quickly compensated.Citation104 Another similar study from the same research group found that resistant populations of Bacteroides persisted at least 2 years after antibiotic administration.Citation105 Resistant Staphylococcus epidermidis have been found 4 years after antibiotic treatment and resistant Enterococcus after 3 years.Citation106,Citation107 These and other studies suggest that even with the reduced usage or absence of antibiotics that antibiotic resistance determinants can be stably maintained in the bacterial population.Citation108

Conclusion

Horizontal gene transfer in the gastrointestinal tract is of particular importance since antibiotic resistance genes can be acquired through transient bacteria or generated through mutations or rearrangements in a variety of elements in this environment. The intestinal microflora is a potential source of antibiotic resistant pathogen. These pathogens can cause infections via the fecal–oral route or through nosocomial infections such as bacteremia, endocarditis, urinary tract infections, and contamination of surgical sites.

The human gastrointestinal tract is a reservoir of enormous species diversity and density as well as a reservoir for hundreds to thousands of known antibiotic resistance genes with the mechanisms in place to horizontally transfer any gene. The capability of emergence of resistant pathogens from this habitat is astounding. The cure for the crisis of antibiotic resistance will not be as simple as encouraging and implementing the judicious use of antibiotics in agriculture and medicine, since resistant strains are very well adapted to growth. It is still unknown how long it will take and if ever resistance gene determinants will be lost from the populations of trillions of bacterial cells that reside in human gastrointestinal tracts worldwide.

Acknowledgments

Work on horizontal gene transfer in the author’s laboratory has been supported by Pursuit Research Grants, Math-Science Grants, and Undergraduate Research Grants at Abilene Christian University.

Disclosure

The author reports no conflicts of interest in this work.

References

  • LevySBMarshallBAntibacterial resistance worldwide: causes, challenges and responsesNat Med200410Suppl 12S122S12915577930
  • de KrakerMEADaveyPGGrundmannHBURDEN study groupMortality and hospital stay associated with resistant Staphylococcus aureus and Escherichia coli bacteremia: estimating the burden of antibiotic resistance in EuropePLoS Med2011810e100110422022233
  • ArmstrongGLConnLAPinnerRWTrends in infectious disease mortality in the United States during the 20th centuryJAMA1999281161669892452
  • SpellbergBPowersJHBrassEPMillerLGEdwardsJEJrTrends in antimicrobial drug development: Implications for the futureClin Infect Dis20043891279128615127341
  • ArutyunovDFrostLSF conjugation: back to the beginningPlasmid2013701183223632276
  • SmillieCGarcillán-BarciaMPFranciaMVRochaEPCde la CruzFMobility of plasmidsMicrobiol Mol Biol Rev MMBR2010743434452
  • ZechnerELLangSSchildbachJFAssembly and mechanisms of bacterial type IV secretion machinesPhilos Trans R Soc B Biol Sci2012367159210731087
  • BaconDJAlmRABurrDHInvolvement of a plasmid in virulence of Campylobacter jejuni 81–176Infect Immun20006884384439010899834
  • WaldorMKTschapeHMekalanosJJA new type of conjugative transposon encodes resistance to sulfamethoxazole, trimethoprim, and streptomycin in Vibrio cholerae O139J Bacteriol199617814415741658763944
  • ZechnerELde la CruzFEisenbrandtRConjugative-DNA transfer processesThomasCMHorizontal Gene Pool: Bacterial Plasmids and Gene SpreadCRC Press200387174
  • GuglielminiJde la CruzFRochaEPEvolution of conjugation and type IV secretion systemsMol Biol Evol201330231533122977114
  • SalyersAAShoemakerNBStevensAMLiLYConjugative transposons: an unusual and diverse set of integrated gene transfer elementsMicrobiol Rev19955945795908531886
  • WalshTRCombinatorial genetic evolution of multiresistanceCurr Opin Microbiol20069547648216942901
  • HallRMIntegrons and gene cassettes: hotspots of diversity in bacterial genomes: HallAnn N Y Acad Sci201212671717822954219
  • BeaberJWHochhutBWaldorMKGenomic and functional analyses of SXT, an integrating antibiotic resistance gene transfer element derived from Vibrio choleraeJ Bacteriol2002184154259426912107144
  • DaviesJDaviesDOrigins and evolution of antibiotic resistanceMicrobiol Mol Biol Rev MMBR2010743417433
  • NormanAHansenLHSorensenSJConjugative plasmids: vessels of the communal gene poolPhilos Trans R Soc B Biol Sci2009364152722752289
  • LorenzMGWackernagelWBacterial gene transfer by natural genetic transformation in the environmentMicrobiol Rev19945835636027968924
  • JohnstonCMartinBFichantGPolardPClaverysJ-PBacterial transformation: distribution, shared mechanisms and divergent controlNat Rev Microbiol201412318119624509783
  • JohnsborgOEldholmVHåvarsteinLSNatural genetic transformation: prevalence, mechanisms and functionRes Microbiol20071581076777817997281
  • SparlingPFGenetic transformation of Neisseria gonorrhoeae to streptomycin resistanceJ Bacteriol1966925136413714958881
  • FinkelSEKolterRDNA as a nutrient: Novel role for bacterial competence gene homologsJ Bacteriol2001183216288629311591672
  • RedfieldRJGenes for breakfast: the have-your-cake-and-eat-it-too of bacterial transformationJ Hered19938454004048409360
  • KristensenBMSinhaSBoyceJDBojesenAMMellJCRedfieldRJNatural transformation of Gallibacterium anatisAppl Environ Microbiol201278144914492222582057
  • ChandlerMSThe gene encoding cAMP receptor protein is required for competence development in Haemophilus influenzae RdProc Natl Acad Sci U S A1992895162616301542653
  • HuddlestonJRBrokawJMZakJCJeterRMNatural transformation as a mechanism of horizontal gene transfer among environmental Aeromonas speciesSyst Appl Microbiol201336422423423541366
  • SinhaSMellJRedfieldRThe availability of purine nucleotides regulates natural competence by controlling translation of the competence activator SxyMol Microbiol20138861106111923663205
  • MichodREWojciechowskiMFHoelzerMADNA repair and the evolution of transformation in the bacterium Bacillus subtilisGenetics1988118131398608929
  • PrudhommeMAttaiechLSanchezGMartinBClaverysJ-PAntibiotic stress induces genetic transformability in the human pathogen Streptococcus pneumoniaeScience20063135783899216825569
  • VosMWhy do bacteria engage in homologous recombination?Trends Microbiol200917622623219464181
  • BacherJMMetzgarDCrécy-LagardV deRapid evolution of diminished transformability in Acinetobacter baylyiJ Bacteriol2006188248534854217028281
  • EngelstädterJMoradigaravandDAdaptation through genetic time travel? Fluctuating selection can drive the evolution of bacterial transformationProc Biol Sci201428117752013260924285199
  • JohnsenPJDubnauDLevinBREpisodic selection and the maintenance of competence and natural transformation in Bacillus subtilisGenetics200918141521153319189946
  • HaijemaBJHahnJHaynesJDubnauDA ComGA-dependent checkpoint limits growth during the escape from competenceMol Microbiol2001401526411298275
  • BiggerJTreatment of staphylococcal infections with penicillin by intermittent sterilisation IThe Lancet19442446320497500
  • LewisKPersister cells, dormancy and infectious diseaseNat Rev Microbiol200751485617143318
  • BakkaliMCould DNA uptake be a side effect of bacterial adhesion and twitching motility?Arch Microbiol2013195427928923381940
  • AverhoffBDNA transport and natural transformation in mesophilic and thermophilic bacteriaJ Bioenerg Biomembr2004361253315168607
  • ChenIDubnauDDNA transport during transformationFront Biosci J Virtual Libr20038s544s556
  • BeikoRGHarlowTJRaganMAHighways of gene sharing in prokaryotesProc Natl Acad Sci U S A200510240143321433716176988
  • FortierL-CSekulovicOImportance of prophages to evolution and virulence of bacterial pathogensVirulence20134535436523611873
  • MuniesaMColomer-LluchMJofreJCould bacteriophages transfer antibiotic resistance genes from environmental bacteria to human-body associated bacterial populations?Mob Genet Elem201334e25847
  • SchmiegerHSchicklmaierPTransduction of multiple drug resistance of Salmonella enterica serovar typhimurium DT104FEMS Microbiol Lett199917012512569919675
  • MannBASlauchJMTransduction of low-copy number plasmids by bacteriophage P22Genetics199714624474569177996
  • DinsdaleEAEdwardsRAHallDFunctional metagenomic profiling of nine biomesNature2008452718762963218337718
  • Mazaheri Nezhad FardRBartonMDHeuzenroederMWBacteriophage-mediated transduction of antibiotic resistance in enterococci: Transduction in Enterococcus sppLett Appl Microbiol201152655956421395627
  • WhitmanWBColemanDCWiebeWJProkaryotes: The unseen majorityProc Natl Acad Sci U S A19989512657865839618454
  • EckburgPBBikEMBernsteinCNDiversity of the human intestinal microbial floraScience200530857281635163815831718
  • LeyREPetersonDAGordonJIEcological and evolutionary forces shaping microbial diversity in the human intestineCell2006124483784816497592
  • SørensenSJBaileyMHansenLHKroerNWuertzSStudying plasmid horizontal transfer in situ: a critical reviewNat Rev Microbiol20053970071016138098
  • LichtTRChristensenBBKrogfeltKAMolinSPlasmid transfer in the animal intestine and other dynamic bacterial populations: the role of community structure and environmentMicrobiology199914592615262210517615
  • KrogfeltKAPoulsenLKMolinSIdentification of coccoid Escherichia coli BJ4 cells in the large intestine of streptomycin-treated miceInfect Immun19936112502950348225579
  • PoulsenLKLanFKristensenCSHobolthPMolinSKrogfeltKASpatial distribution of Escherichia coli in the mouse large intestine inferred from rRNA in situ hybridizationInfect Immun19946211519151947927805
  • MarksLRMashburn-WarrenLFederleMJHakanssonAPStreptococcus pyogenes biofilm growth in vitro and in vivo and its role in colonization, virulence and genetic exchangeJ Infect Dis2014 Epub January 23, 2014
  • MillsSShanahanFStantonCHillCCoffeyARossRPMovers and shakersGut Microbes20134141623022738
  • AnwarHDasguptaMKCostertonJWTesting the susceptibility of bacteria in biofilms to antibacterial agentsAntimicrob Agents Chemother19903411204320462073094
  • SalyersAAGuptaAWangYHuman intestinal bacteria as reservoirs for antibiotic resistance genesTrends Microbiol200412941241615337162
  • LiuBPopMARDB – Antibiotic Resistance Genes DatabaseNucleic Acids Res200937suppl 1D443D44718832362
  • GhoshTSGuptaSSNairGBMandeSSIn silico analysis of antibiotic resistance genes in the gut microflora of individuals from diverse geographies and age-groupsPLoS ONE2013812e8382324391833
  • SommerMOADantasGChurchGMFunctional characterization of the antibiotic resistance reservoir in the human microfloraScience200932559441128113119713526
  • HuYYangXQinJMetagenome-wide analysis of antibiotic resistance genes in a large cohort of human gut microbiotaNat Commun20134215123877117
  • ForslundKSunagawaSKultimaJRCountry-specific antibiotic use practices impact the human gut resistomeGenome Res20132371163116923568836
  • MarshallBMLevySBFood animals and antimicrobials: impacts on human healthClin Microbiol Rev201124471873321976606
  • SchjorringSusanneKrogfeltKAAssessment of bacterial antibiotic resistance transfer in the gutInt J Microbiol20112011110
  • CarrFJChillDMaidaNThe lactic acid bacteria: A literature surveyCrit Rev Microbiol200228428112546196
  • AmmorMSFlórezABMayoBAntibiotic resistance in non-enterococcal lactic acid bacteria and bifidobacteriaFood Microbiol200724655957017418306
  • PerretenVSchwarzFCrestaLBoeglinMDasenGTeuberMAntibiotic resistance spread in foodNature199738966538018029349809
  • EgervärnMLindmarkHOlssonJRoosSTransferability of a tetracycline resistance gene from probiotic Lactobacillus reuteri to bacteria in the gastrointestinal tract of humansAntonie Van Leeuwenhoek201097218920019997864
  • DragoLRodighieroVMattinaRToscanoMDE VecchiEIn vitro selection and transferability of antibiotic resistance in the probiotic strain Lactobacillus reuteri DSM 17938J Chemother Florence Italy2011236371373
  • LesterCHFrimodt-MøllerNSørensenTLMonnetDLHammerumAMIn vivo transfer of the vanA resistance gene from an Enterococcus faecium isolate of animal origin to an E. faecium isolate of human origin in the intestines of human volunteersAntimicrob Agents Chemother200650259659916436715
  • TrobosMLesterCHOlsenJEFrimodt-MøllerNHammerumAMNatural transfer of sulphonamide and ampicillin resistance between Escherichia coli residing in the human intestineJ Antimicrob Chemother2009631808618957394
  • CrémetLBourigaultCLepelletierDNosocomial outbreak of carbapenem-resistant Enterobacter cloacae highlighting the interspecies transferability of the blaOXA-48 gene in the gut floraJ Antimicrob Chemother20126741041104322223227
  • KaramiNMartnerAEnneVISwerkerssonSAdlerberthIWoldAETransfer of an ampicillin resistance gene between two Escherichia coli strains in the bowel microbiota of an infant treated with antibioticsJ Antimicrob Chemother20076051142114517768176
  • GorenMGCarmeliYSchwaberMJChmelnitskyISchechnerVNavon-VeneziaSTransfer of carbapenem-resistant plasmid from Klebsiella pneumoniae ST258 to Escherichia coli in patientEmerg Infect Dis20101661014101720507761
  • Duval-IflahYRaibaudPTancredeCRousseauMR-plasmic transfer from Serratia liquefaciens to Escherichia coli in vitro and in vivo in the digestive tract of gnotobiotic mice associated with human fecal floraInfect Immun19802839819906995330
  • SparoMUrbizuLSolanaMVHigh-level resistance to gentamicin: genetic transfer between Enterococcus faecalis isolated from food of animal origin and human microbiotaLett Appl Microbiol201254211912522098378
  • LesterCHFrimodt-MollerNHammerumAMConjugal transfer of aminoglycoside and macrolide resistance between Enterococcus faecium isolates in the intestine of streptomycin-treated miceFEMS Microbiol Lett2004235238539115183889
  • CarattoliAResistance plasmid families in EnterobacteriaceaeAntimicrob Agents Chemother20095362227223819307361
  • KhajuriaAPraharajAKKumarMGroverNAggarwalAMultidrug resistant NDM-1 metallo-beta-lactamase producing Klebsiella pneumoniae sepsis outbreak in a neonatal intensive care unit in a tertiary care center at central IndiaIndian J Pathol Microbiol2014571656824739834
  • SchlüterANordmannPBonninRAIncH-type plasmid harboring the bla CTX-M-15, bla DHA-1, and qnr B4 genes recovered from animal isolatesAntimicrob Agents Chemother2014
  • MarchaimDChopraTPogueJMOutbreak of colistin-resistant, carbapenem-resistant Klebsiella pneumoniae in Metropolitan Detroit, MichiganAntimicrob Agents Chemother2011552593599 [Epub ahead of print]21115786
  • LongKSPoehlsgaardJKehrenbergCSchwarzSVesterBThe Cfr rRNA methyltransferase confers resistance to phenicols, lincosamides, oxazolidinones, pleuromutilins, and streptogramin A antibioticsAntimicrob Agents Chemother20065072500250516801432
  • De VriesLEVallèsYAgersøYThe gut as reservoir of antibiotic resistance: Microbial diversity of tetracycline resistance in mother and infantPLoS ONE201166e2164421738748
  • ShoemakerNBVlamakisHHayesKSalyersAAEvidence for extensive resistance gene transfer among Bacteroides spp. and among Bacteroides and other genera in the human colonAppl Environ Microbiol200167256156811157217
  • RadmanMSOS repair hypothesis: phenomenology of an inducible DNA repair which is accompanied by mutagenesisBasic Life Sci19755A3553671103845
  • MichelBAfter 30 years of study, the bacterial SOS response still surprises usPLoS Biol200537e25516000023
  • VarhimoESavijokiKJefremoffHJalavaJSukuraAVarmanenPCiprofloxacin induces mutagenesis to antibiotic resistance independent of UmuC in Streptococcus uberisEnviron Microbiol20081082179218318430157
  • AlonsoACampanarioEMartínezJLEmergence of multidrug-resistant mutants is increased under antibiotic selective pressure in Pseudomonas aeruginosaMicrobiology1999145102857286210537207
  • KimJNohJParkWInsight into norfloxacin resistance of Acinetobacter oleivorans DR1: target gene mutation, persister, and RNA-Seq analysesJ Microbiol Biotechnol20132391293130323928845
  • CharpentierXKayESchneiderDShumanHAAntibiotics and UV radiation induce competence for natural transformation in Legionella pneumophilaJ Bacteriol201119351114112121169481
  • WhitchurchCBTolker-NielsenTRagasPCMattickJSExtracellular DNA required for bacterial biofilm formationScience20022955559148711859186
  • CharpentierXPolardPClaverysJ-PInduction of competence for genetic transformation by antibiotics: convergent evolution of stress responses in distant bacterial species lacking SOS?Curr Opin Microbiol201215557057622910199
  • LemireSFigueroa-BossiNBossiLBacteriophage crosstalk: Coordination of prophage induction by trans-acting antirepressorsPLoS Genet201176e100214921731505
  • UlrichRLDeshazerDKennyTACharacterization of the Burkholderia thailandensis SOS response by using whole-transcriptome shotgun sequencingAppl Environ Microbiol201379195830584323872555
  • ModiSRLeeHHSpinaCSCollinsJJAntibiotic treatment expands the resistance reservoir and ecological network of the phage metagenomeNature2013499745721922223748443
  • CollinsSMDenouEVerduEFBercikPThe putative role of the intestinal microbiota in the irritable bowel syndromeDig Liver Dis Off J Ital Soc Gastroenterol Ital Assoc Study Liver20094112850853
  • ReiffCKellyDInflammatory bowel disease, gut bacteria and probiotic therapyInt J Med Microbiol IJMM201030012533
  • WagnerJMaksimovicJFarriesGBacteriophages in gut samples from pediatric Crohn’s disease patients: metagenomic analysis using 454 pyrosequencingInflamm Bowel Dis20131981598160823749273
  • BeaberJWHochhutBWaldorMKSOS response promotes horizontal dissemination of antibiotic resistance genesNature20044276969727414688795
  • ÚbedaCMaiquesEKnechtELasaÍNovickRPPenadésJRAntibiotic-induced SOS response promotes horizontal dissemination of pathogenicity island-encoded virulence factors in staphylococci: Horizontal dissemination of virulence genesMol Microbiol200556383684415819636
  • AnderssonDILevinBRThe biological cost of antibiotic resistanceCurr Opin Microbiol19992548949310508723
  • BjorkmanJHughesDAnderssonDIVirulence of antibiotic-resistant Salmonella typhimuriumProc Natl Acad Sci U S A1998957394939539520473
  • BakerSDuyPTNgaTVTFitness benefits in fluoroquinolone-resistant Salmonella typhi in the absence of antimicrobial pressureeLife20132
  • LöfmarkSJernbergCBillströmHAnderssonDIEdlundCRestored fitness leads to long-term persistence of resistant Bacteroides strains in the human intestineAnaerobe200814315716018434218
  • JernbergCLöfmarkSEdlundCJanssonJKLong-term ecological impacts of antibiotic administration on the human intestinal microbiotaISME J200711566618043614
  • SjölundMTanoEBlaserMJAnderssonDIEngstrandLPersistence of resistant Staphylococcus epidermidis after single course of clarithromycinEmerg Infect Dis20051191389139316229767
  • SjölundMWreiberKAnderssonDIBlaserMJEngstrandLLong-term persistence of resistant Enterococcus species after antibiotics to eradicate Helicobacter pyloriAnn Intern Med2003139648348713679325
  • JakobssonHEJernbergCAnderssonAFSjölund-KarlssonMJanssonJKEngstrandLShort-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiomePloS One201053e983620352091