Abstract
Antibiotic-mediated perturbation of the gut microbiome is associated with numerous infectious and autoimmune diseases of the gastrointestinal tract. Yet, as the gut microbiome is a complex ecological network of microorganisms, the effects of antibiotics can be highly variable. With the advent of multi-omic approaches for systems-level profiling of microbial communities, we are beginning to identify microbiome-intrinsic and microbiome-extrinsic factors that affect microbiome dynamics during antibiotic exposure and subsequent recovery. In this Review, we discuss factors that influence restructuring of the gut microbiome on antibiotic exposure. We present an overview of the currently complex picture of treatment-induced changes to the microbial community and highlight essential considerations for future investigations of antibiotic-specific outcomes. Finally, we provide a synopsis of available strategies to minimize antibiotic-induced damage or to restore the pretreatment architectures of the gut microbial community.
Introduction
The human gut microbiome is a microbial organ with substantial influence over health and disease1. From birth, the gut microbiome works in cohesion with the gut epithelium and surrounding organs to digest and transform nutrients, provide colonization resistance against pathogens and regulate the immune system2–4. Consequently, disruptions in the taxonomic and metabolic pathway architecture of the gut microbial community (referred to as dysbiosis) have been associated with an increased risk of numerous human pathologies, including inflammatory bowel disease5, obesity6, colorectal cancer7,8 and numerous gastrointestinal infections9–11. Among the most notable aetiologies of gut microbiome dysbiosis is antibiotic exposure. In this Review, we focus on the relationship between antibiotics and the human gut microbiome, the complexities of understanding how antibiotics acutely and persistently perturb this microbial community, and strategies for remediating these perturbations.
Since the discovery of penicillin by Alexander Fleming12, antibiotics have revolutionized modern medicine, saving countless lives from otherwise lethal bacterial infections13. For instance, in the USA, the antibiotic revolution changed the leading causes of death from infectious diseases (for example, pneumonia, tuberculosis or diphtheria) to non-communicable diseases (for example, cardiovascular diseases or cancer), adding decades to the average life expectancy13,14. Thus, antibiotics have critically enabled our modern ways of living, particularly in high-income countries. However, although antibiotics were developed to target human and animal pathogens of interest, their molecular targets (for example, the cell wall, the ribosome and RNA polymerase) are highly conserved across the bacterial kingdom. Accordingly, antibiotic exposure in the gut can indiscriminately target both pathogenic and benign bacteria, thereby disrupting ecological niches that are responsible for myriad metabolic transformations. This change in taxonomic composition of the gut microbiome is often a risk factor for the onset of other diseases. Most notably, treatment-induced disturbance of the gut community enables the growth of potentially infectious pathobionts (opportunistic microorganisms that proliferate on microbiome perturbation) with the capacity to cause disease15,16. Some of the most dangerous microbial threats (as named by the CDC in 2019 (ref. 17)), including drug-resistant Enterobacteriaceae, Clostridioides difficile, vancomycin-resistant Enterococcus and Salmonella spp., are responsible for a substantial amount of gastrointestinal disease18. In addition to enrichment of overtly pathogenic organisms following antibiotic exposure, antibiotics are increasingly considered key contributors to more complex immunological manifestations such as graft-versus-host disease, inflammatory bowel disease and even allergies in the host5,19–21. Understanding how different antibiotics disturb the human gut microbiome is thus critical for mitigating diseases related to microbiome dysbiosis.
Another unintended consequence of the steady increase in antibiotic utilization worldwide22 has been the concomitant rise of antimicrobial resistance (AMR), compromising the treatment of resistant bacterial infections23,24. In 2019, an estimated 4.95 million deaths were associated with bacterial AMR worldwide25. Enrichment for AMR often begins within the gut microbiome of the patient, with antibiotics increasing the abundance of resident bacteria that carry antibiotic resistance genes (ARGs) and altering ARG content within the gut microbiome (termed the ‘resistome’)26,27. However, the spread of ARGs is not exclusively attributed to their primary bacterial hosts: ARGs are commonly encoded within mobile genetic elements (MGEs)28, which enable their acquisition by new hosts, including pathobionts29. Notably, the presence of ARG-encoding pathobionts in the gut is a risk factor for recurrent resistant infections post-treatment30. Knowledge regarding the impact of specific antibiotics on gut resistome structure and dynamics, with a particular focus on mobilizable ARGs, could guide future treatment choices, leading to increased therapeutic efficacy long-term and minimizing the dissemination of AMR in the gut microbiome.
In this Review, we incorporate primary literature describing human cohort studies, animal studies and in vitro bacteriology to provide an overview of the factors governing the antibiotic exposure-induced restructuring of the gut microbial community. In each case, we highlight the general patterns of change in bacterial populations and encoded resistance traits observed during treatment, while emphasizing the multiple levels of complexity in understanding antibiotic action in a community of diverse, interconnected microorganisms within the human host. Finally, we review promising therapeutic avenues aimed at minimizing antibiotic-induced damage to the microbiome or restoring the community structure post-treatment.
Effects on the gut microbiome
A growing body of research has investigated the structure of the gut microbiome and its dynamics after different types of antimicrobial exposures (Supplementary Table 1). Antibiotic-specific effects on the gut microbiome are increasingly recognized, and both human and animal studies have revealed reproducible taxonomic and metabolic patterns of community perturbation, particularly in the context of broad-spectrum therapies. In this section, we first present general trends observed on antibiotic administration. Subsequently, to illustrate antibiotic-specific effects on the gut microbiome, we rely on the results of a few human clinical trials comparing the effects of distinct antibiotics on the microbiome in a controlled manner (Supplementary Table 1). Last, we highlight the growth and metabolic characteristics of bacterial life cycles in the gastrointestinal tract that alter bacterial resiliency during antibiotic treatment.
Generalized and antibiotic-specific principles of microbiome community perturbation
In general, exposure to antibiotics (across drug classes) often causes important changes in microbiome community structure, species composition and metabolic capacity (Fig. 1). Foundational studies in healthy adult humans have commonly demonstrated the acute decline in alpha diversity31 (in the first couple of days of treatment) and the incomplete recovery of the microbiome up to 6 months after treatment (often measured by beta diversity between baseline and post-treatment)32–37. These community-level changes are accompanied by significant decreases in the relative abundance of important members of the phyla Firmicutes, Bacteroidetes and Actinobacteria, most notably Faecalibacterium prausnitzii, Eubacterium spp., Roseburia spp., Bifidobacterium spp., Dorea spp., Anaerostipes spp. and Ruminococcus spp.32,34,38,39. These lost species provide critical metabolic functions to the gut microbiome, as antibiotic administration in both humans and mice is commonly associated with decreased bile acid transformation capacity (increased primary bile acids) and carbohydrate fermentation (decreased short-chain fatty acids (SCFAs))40–45. Additionally, metabolomic investigations of antibiotic treatment and the gut microbiome have noted increased levels of simple carbohydrates and amino acids, which is probably due to the loss of species diversity in the community45,46. Loss of this metabolic colonization resistance through antibiotic treatment precedes opportunistic infections (or blooms if pathobionts were existent at a lower abundance) by C. difficile, Enterococcus spp., Candida albicans and other gut pathogens46–50. Correspondingly, antibiotic treatment in adult humans is often associated with an increase in the relative abundance of facultative anaerobes that were initially less abundant members of the community, such as Enterobacteriaceae, Enterococcus spp., Clostridium spp. and Streptococcus spp.34,37,51,52. Although clinical studies of bacterial infections often emphasize the species and/or strain of opportunistic pathogens or pathobionts53,54, literature describing commensal fluctuations during antibiotic exposure is currently limited to the taxonomic resolution of genus or species.
Fig. 1|. Antibiotic-mediated destruction of the gut microbiome opens an opportunistic niche.

Antibiotics often eradicate Bacteroidetes, Firmicutes and Actinobacteria taxa responsible for important gut microbiome functions, although the species lost are community-specific and antibiotic-specific. This loss of diversity leaves an open niche for colonization or proliferation by opportunistic bacteria that are associated with gastrointestinal disease, namely those from phylum Proteobacteria. These opportunistic bacteria often have mobilizable drug resistance or innate properties such as spore formation that enable them to survive antibiotic exposure. A number of organisms that can colonize an antibiotic-perturbed gut microbiome are recognized by the CDC as major threats to public health. ESBL, extended-spectrum β-lactamase.
Assessment of gut microbiome composition is confounded by numerous host, microbial and environmental factors (both technical and biological), rendering comparisons of the reported effects of different antibiotics on the gut across studies currently intractable. Our understanding of the differential impact of antibiotics is largely informed by investigations in which multiple treatment agents are analysed in the same study population39,46,55,56. For instance, vancomycin has been shown to cause a significant decrease in alpha diversity (more than twofold at the operational taxonomic unit level), an increase in primary bile acids (approximately threefold in faecal levels) and a decrease in the stool butyrate and acetate levels (more than fivefold and approximately twofold, respectively) relative to amoxicillin39,55; paradoxically, of the two drugs, vancomycin has a narrower in vitro spectrum of activity. Furthermore, vancomycin significantly decreased the relative abundance of Coprococcus eutactus, F. prausnitzii and Anaerostipes caccae (SCFA-producing Firmicutes), while increasing the relative abundance of Enterobacteriaceae and Enterococcus spp.55. Three different mouse models have reproduced the superior capacity of vancomycin to decrease alpha diversity or increase pathogen susceptibility compared to more than five other antibiotic agents or combinations (including ampicillin, azithromycin, ciprofloxacin and ciprofloxacin– metronidazole)46,56,57. Relative to cotrimoxazole and nitrofurantoin, ciprofloxacin has been shown to cause a greater reduction in species richness (less than twofold) and beta diversity58,59, in addition to its effect in depleting Bifidobacterium and Ruminococcus spp. Azithromycin has been widely explored (in clinical trials of >1,000 children across sub-Saharan Africa) on its own and in comparison to other antibiotics60–66. In a comparative study, azithromycin leads to a greater decrease in Simpson’s alpha diversity (less than twofold) 5 days post-treatment than amoxicillin or cotrimoxazole67. More recently, in healthy adult volunteers, microbiome analysis during four different antibiotic regimens identified that azithromycin-containing regimens were associated with delayed community recovery following treatment relative to the regimens without azithromycin33. Understanding perturbation differences between regimens will be critical to enhancing antibiotic stewardship, and future studies incorporating more control over antibiotic administration parameters (for example, route of exposure, duration, dose or bioavailability) will be imperative for influencing antibiotic stewardship.
To systematically benchmark magnitudes of community destruction in the microbiome during different antibiotic perturbations, an increased taxonomic resolution of the commensal microbiome and a refined appreciation for antibiotic pharmacodynamics are required. To date, most studies of the microbiome have relied on stool short-read shotgun metagenomics or 16S rRNA amplicon sequencing as the main techniques for determining community structure, which generally does not provide strain-level resolution of the microbiome (a requirement for understanding gut microbiome AMR composition, as discussed below)68,69. Metagenome-assembled genomes, particularly those incorporating long-read metagenomic sequencing, can provide increased strain-level resolution70–72. Additionally, depending on the chemical composition, formulation and administration route, antibiotics have different propensities to bioaccumulate in the gastrointestinal tract73; this undoubtedly has a role in determining the magnitude of microbiome perturbation. For example, the superior ability of oral vancomycin administration to perturb the microbiome may be due to its poor absorption in mammals and perhaps higher concentration in the lumen74. Bioavailability of antibiotics and their administration route have been rarely examined as influential variables in comparative studies of microbiome perturbation75,76. Although published human studies provide some grounds for making antibiotic-specific claims about different magnitudes of microbiome perturbation capacity, human and animal models require more rigorous control over dosing and route of administration of antibiotics to accurately interpret differences in effect size across antibiotics77.
Species and community determinants of susceptibility during antibiotic exposure
From the perspective of an individual species of the gut microbiome, several intrinsic growth properties may enable a subset of bacterial species to persist on antibiotic treatment (Fig. 2). Members of the Clostridium and Peptostreptococcus genera (along with several other species in the phylum Firmicutes) have the capacity to form endospores, which are non-replicating forms of the cell that are intrinsically resistant to antibiotics, heat and oxygen78–80. Spore-forming pathobionts such as C. difficile and Clostridium perfringens sporulate in response to population density and nutrient availability81. These spores survive antibiotic exposure in the gastrointestinal tract, and the presence of free nutrients and primary bile acids (as discussed above) provides molecular signals for spore germination and subsequent pathogenesis82,83. Apart from spore-mediated persistence, many gut microorganisms can tolerate antibiotics through various resiliency mechanisms, including persister cell formation, perhaps in a mucin-embedded biofilm or deeper tissue niche where they can resist antibiotics84–87. Antibiotic susceptibility of a gut commensal may thus be determined by its surrounding nutrient milieu (Box 1), a concept that remains difficult to investigate in vivo88–92. Although it is often speculated that Bacilli and Enterobacteriaceae are more likely to bloom on antibiotic treatment of the gut microbiome because they possess more encoded resistance, these suppositions are based on resistance databases with likely overrepresentation of pathogens (discussed below) and do not account for the growth and metabolism of single organisms and communities.
Fig. 2|. Integrated understanding of how antibiotics remodel the microbiome.

Mechanistic investigation into taxonomic, metabolic and resistome restructuring during antibiotic exposure requires multiple levels of study: single organisms, multi-species dynamics and in vivo microbiome studies in animal and human hosts. The susceptibility of a single organism is governed by physiological properties such as spore-forming capacity, the presence of drug resistance genetic elements, or defined differences in nutrients and minerals. Examination of bacterial interactions has revealed that the surrounding environment can influence differential susceptibility through provocation of horizontal gene transfer, metabolic conditioning or cell–cell signalling. In increasingly complex communities, the metabolic network of cross-feeding interactions imparts unknown consequences on microbial susceptibility (at the species or community level). The exchange of antimicrobial resistance genes or other mobile genetic elements could also drive the outcome of community perturbation. Finally, at the whole-organism level, factors external to the microbial community may determine the magnitude of an antibiotic perturbation. Host genetics and immune status are likely to influence drug metabolism and absorption, in addition to drug-specific properties of absorption. Additionally, dynamic influences such as host consumption of dietary elements and medications are likely to change community trajectories during antibiotic perturbation. Other factors not discussed here are highlighted in Table 1.
Box 1. Examples of metabolite–microorganism–antibiotic dynamics relevant to the gut microbiome.
Host-derived metabolites and bacterial signalling molecules have the capacity to change the intrinsic antibiotic susceptibility of bacterial species. In phylum Bacteroidetes, Bacteroides thetaiotaomicron is more tolerant of amoxicillin in the presence of polysaccharides such as pullulan, whereas glucose consumption is sensitizing88 (see the figure, part a). Human milk oligosaccharides cause changes to membrane permeability, sensitizing Streptococcus agalactiae to trimethoprim89 (see the figure, part b). As this species is not a member of the gut microbiome, it is possible that streptococci and other organisms in the gut experience differences in membrane permeabilization due to fluctuations in dietary polysaccharides. Indole-3-acetic acid, which is a derivative of microbial-derived tryptophan metabolism, can sensitize Staphylococcus aureus to ciprofloxacin90 (see the figure, part c). In phylum Proteobacteria, indole (another microbially produced tryptophan derivative) is produced by Enterobacteriaceae and can induce tolerance to quinolones in nearby Enterobacteriaceae91,92 (see the figure, part d). These examples highlight representative and simple nutrient–microorganism relationships but remain difficult to generalize to the complex community within the gut microbiome.
MRSA, methicillin-resistant S. aureus.
In clinical practice, in vitro susceptibilities for respiratory and enteric pathogens serve as the gold standard for treatment of such infection and predict pathogen killing reasonably well. Yet, the in vitro spectrum of activity of an antibiotic does not always predict in vivo species trajectories in a multi-species microbiome during antibiotic exposure93–96. For example, despite the in vitro Gram-positive-specific action of vancomycin, its oral administration (in human and animals) results in distinct loss of several Gram-negative gut microbiome commensals including Bacteroides, Prevotella and Alistipes spp.97. These observations underline the complexity of microbial metabolic networks in the microbiome, built by a diversity of mutualistic and competitive relationships across phyla. These community networks almost certainly alter antibiotic killing in the microbiome98. In vitro studies of interactions between two gut commensals during antibiotic treatment indicate that symbiotic relationships can have multiple outcomes in effecting differences in antibiotic tolerance in one99 or both members of the community100,101. Although these studies provide a high-resolution view of model gut microbiome relationships, extrapolating these findings to much larger networks of relationships within the human gut microbiome remains a critical hurdle in the field. This phenomenon is further complicated given that different investigations of microbiome perturbation by the same antibiotic often do not agree (Supplementary Table 1).
Host-related contributions
Synergy between antibiotics and metabolic or dietary elements can further shape microbiome dynamics and alter susceptibility to antibiotics within the community, leading to clinically relevant differences in disease102 (Fig. 2). Investigations into ‘Western-style’ diets (high in fat and simple sugars) have revealed that these diets correlate with an increased risk for obesity and heart disease, among other poor health outcomes103,104. Antibiotic treatment of mice on a high-fat diet provokes pathological inflammation at the gut mucosa and increased levels of Enterobacterales, a taxonomic order that contains common pathobionts of the microbiome105. These mice displayed immunological symptoms of pre-inflammatory bowel disease, which is probably due to the combined metabolic influence of the high-fat diet on the gut epithelial cells and the change in microbiome structure. From several other mouse studies using both dietary and chemical interventions to simulate the effects of a Western-style diet, it is clear that the combination of both diet and antibiotics changes microbial and host metabolism106. Specifically, in one model, such synergistic interventions freed up simple sugars and ethanolamines through an altered fatty acid metabolism, ultimately increasing susceptibility to enteric infection by Salmonella enterica107. Alternatively, modulation of polysaccharide or microbiota-accessible carbohydrate levels in diet is highly influential over antibiotic treatment outcomes. Without fibre, mice with a conventional microbiome or a humanized microbiome have a delayed recovery in alpha diversity after ciprofloxacin treatment96. In humans, volunteers fed omnivore and vegan diets during combination antibiotic treatment displayed an expedited microbiome recovery (as measured by alpha and beta diversity) relative to those on a fibre-free liquid diet. Specifically, fibre-containing diets encouraged a rapid restoration of stool butyrate levels and a return of Firmicutes with distinct amino acid metabolism108.
In addition to understanding macronutrient dietary influences on antibiotic perturbations, consumption of antibiotics is likely to be accompanied by consumption of other medications, differential micronutrient intake and differential xenobiotic exposure. The interactions between these non-antibiotic factors and the microbiome have been reviewed elsewhere109. Metagenomic analyses of large cohorts of humans with gastrointestinal inflammatory conditions reveal strong taxonomic and metabolic pathway associations with a proton-pump inhibitor, metformin (a type 2 diabetes medication) and laxative exposure110. These extrinsic host influences are not typically accounted for in antibiotic–microbiome investigations, despite the clear capacity of many of these xenobiotic compounds to affect growth promotion or inhibition in a species-specific manner111,112. Correspondingly, some pharmaceuticals have the capacity to antagonize or synergize with antibiotics. For instance, dicumarol, an anticoagulant, was demonstrated in vitro and in vivo to antagonize the erythromycin-mediated killing of Bacteroides vulgatus, while allowing the killing of the opportunistic pathogen Enterococcus faecalis in a 12-member defined community113. Future human studies on coadministered medications and antibiotics will be critical, especially in immunocompromised hosts, to understanding the risks and benefits of coadministration related to microbiome consequences.
Effects on the gut resistome
Antibiotics have a pronounced but highly variable effect on the gut resistome (the suite of ARGs encoded by the microbiome)33. Here, we summarize key insights from the literature regarding the effects of antibiotics on the resistome, while highlighting factors that correlate with variability in treatment outcomes. Although resistome composition is intimately connected to the taxonomic structure of the gut microbiome (defined by a distinct population of bacterial strains encoding resistance-conferring elements)52, our increasing understanding of horizontal gene transfer (HGT) has revealed that this connection is not absolute and that changes in the resistome are not fully explained by concurrent taxonomic shifts. In this context we focus on HGT within the microbiome, discussing the potential roles of MGEs in the spread of ARGs post-antibiotic challenge and emphasizing the challenges accompanying the investigations into the impact of antibiotics on HGT.
Antibiotics commonly enrich the gut resistome
Changes to the gut resistome through antibiotic exposure could provoke pathological blooms of drug-resistant pathobionts in the microbiome. Because of these phenomena, the composition and dynamics of the gut resistome have been the foci of numerous studies (Table 1), which have revealed a few general patterns of antibiotic-induced changes in the resistome. Notably, antibiotic treatment most commonly results in acutely increased ARG burden (as measured by changes in relative abundance) in the gut microbiome33,51,114–118. Despite increasing the total resistome burden, antibiotics can concurrently lead to a substantial loss of specific ARGs119 and reduced resistome diversity116, which is probably due to taxonomic loss. Antibiotics expectedly enrich for ARGs that confer resistance to the treatment agent33,34,51,62–66,114,115,117,119–123 through selection of bacterial strains encoding these genes121,124,125. However, concurrent enrichment of ARGs unrelated to the administered drug is common33,34,51,114,115,119–121,123. This latter enrichment of ARGs after antibiotics is often species-specific, and the set of ARGs enriched on treatment can be attributed to specific multi-drug resistant (MDR) taxa52,126. For example, the enrichment of most ARGs in the guts of preterm infants treated with meropenem and ticarcillin–clavulanate was found to be highly correlated with the enrichment of Staphylococcus epidermidis or Klebsiella pneumoniae52. However, it should be noted that ARG carriage is a strain-specific trait, and it is likely that individual MDR traits within these species are being selected for124. Another factor driving the enrichment of genes unrelated to the treatment is the localization of diverse ARGs within MDR genetic clusters, which ensures the co-enrichment of groups of genes on selection for at least one of the encoded ARGs52,127. However, such co-selection of ARGs is often short-lived: with prolonged treatment, ARGs conferring resistance to the administered antibiotic remain enriched even after years, but the abundance of enriched ARGs unrelated to treatment can diminish with time62. This suggests that over time, microorganisms undergo more specialized adaptation, isolating and maintaining the ARGs conferring growth advantage to the host while putatively shedding the genes unrelated to the treatment (likely due to the fitness costs associated with carriage of some ARGs)128.
Table 1|.
Biological variables that influence gut microbiome dynamics in response to antibiotics
| Variable | Example | Refs. |
|---|---|---|
| Antibiotic type or class | Azithromycin (macrolide) versus levofloxacin (fluoroquinolone) | 33,39,52,123,126,210–214 |
| Spectrum of activity | Broad versus narrow spectrum | 55 |
| Drug pharmacokinetics | Biliary elimination rate | 215 |
| Administration route | Oral versus intravenous | 125,216 |
| Antibiotic dose | Low-dose versus high-dose treatment | 59,211,217 |
| Duration of treatment | Short-term versus prolonged treatment | 62–66,218 |
| Concurrent medication or medications | Virostatic agents | 59 |
| Baseline microbiome composition | Abundance of Bacteroides | 59,68,219 |
| Diet | Low-fibre diet; breast milk | 52,96,108,220 |
| Host age | Postmenstrual age | 52,126 |
| Health status | Clinical Risk Index for Babies (CRIB) II score | 52 |
| Host organ system activity | Kidney and liver function | 59 |
| Sex | Male versus female | 56,221,222 |
| Host genetics | C57BL/6J mice versus 129S6 mice | 223 |
| Host type | Drug clearance rate in humans versus mice | 224 |
Importantly, the effects of antibiotics on the gut resistome vary substantially depending on the nature of the administered drug33,52,59,114,123. For instance, in one study involving healthy adults, oral administration of azithromycin (a macrolide) or cefpodoxime (a cephalosporin) resulted in an increased relative abundance of ARGs, whereas no changes in the relative ARG abundance were observed on oral administration of levofloxacin (a fluoroquinolone)33. Similar post-treatment effects of macrolides62–66,123, cephalosporins119,120 and fluoroquinolones59 on the relative abundance of ARGs have been reported elsewhere for diverse population segments (for example, children and elderly, healthy and diseased). Nonetheless, the effects of various antibiotic classes and individual antibiotics on the human gut resistome remain largely unexplored; reports focusing on single-treatment agents remain few and far between33,59,62,119–121,123, with investigations of the resistome changes commonly involving combinations of antibiotics32,34,122 or with retrospective cohorts with variable treatment regimens, doses and durations114,116,118,129 (Table 1). Moreover, factors beyond the treatment agent can affect resistome dynamics during treatment, further obscuring our understanding of the antibiotic-induced resistome changes. Notably, administration route is a potentially important determinant of the extent of resistome restructuring by antibiotics. In mice treated with tetracycline or ampicillin, relative to intravenous injection, oral administration of the same doses of drugs resulted in faster and higher increases in relative abundance of ARGs125. To achieve a more in-depth understanding of antibiotic-specific effects on the gut resistome, there is a need for future research involving single-treatment agents tested within the same study populations with controls over dose and administration routes.
To use our nascent understanding of the antibiotic-mediated changes to the resistome in any predictive manner, the field would benefit from establishment and broader implementation of standardized approaches for resistome characterization. Recent investigations of the gut resistome have primarily relied on whole-metagenome shotgun sequencing with subsequent alignment of sequencing reads or annotated open reading frames from assembled contigs against one or a few of the available databases of known ARGs32–34,59,62,114,117–119,123,129,130. ARG databases vary considerably in size (that is, the number of represented resistance determinants) and composition131, and, consequently, resistome classification is not consistent across databases132. There are instances of the same gene annotated by different names across databases133, which further impedes accurate comparative analysis of corresponding resistome studies. Furthermore, such databases are typically curated from the existing biomedical literature134,135, which suggests a strong bias in these collections towards ARGs from human pathogens and underrepresentation of ARGs from commensals and environmental organisms. These shortcomings could be addressed through functional metagenomic screens, which identify novel ARGs in a sequence-agnostic, function-centric manner136. Future efforts to establish the effects of antibiotics on the gut resistome would benefit from enrichment of antimicrobial resistance databases with ARGs from a broader range of organisms and habitats, and from ongoing consolidation, cross-checking and benchmarking of these databases.
Horizontal gene transfer facilitates post-antibiotic resistome enrichment
The antibiotic-induced expansion of the gut resistome may also be driven by ARG-encoding MGEs114,137,138. In the short term, the increase in resistome burden after antibiotic treatment is primarily driven by the enrichment of chromosomally encoded ARGs (that is, the expansion of resistant bacterial lineages). However, the abundance of microorganisms with chromosomal ARGs declines sharply shortly thereafter (~1 month)68. Conversely, MGE-encoded ARGs persist for longer periods after therapy cessation, which is probably due to the parasitic nature of MGEs (that is, their persistence among bacterial populations despite the absence of antibiotic-mediated growth advantages or presence of fitness defects conferred on the bacterial host)32,68. Antibiotics can increase the abundance of MGE-encoded ARGs in the gut59 and lead to more frequent and broader (involving more diverse taxa) dissemination of ARGs and MGEs in patients138. Overall, higher rates of horizontal transfer of ARGs are found in populations with higher reported clinical use and environmental antibiotic exposure139. Such increased HGT rates are of concern owing to their potential implication in the spread of ARGs to both resident commensals and pathobionts within the gut microbiome140, thus increasing the future risk of opportunistic infections.
Antibiotics can reportedly facilitate all three mechanisms of HGT: transformation141–143, transduction144 and conjugation145,146, the last being the best-studied form of horizontal transfer of ARGs138,147–152 (Box 2). ARGs are commonly encoded on conjugative plasmids, which can be transferred between taxonomically distinct residents of the gut microbiome137. Low doses of antibiotics have been reported to facilitate plasmid conjugation within bacterial communities145,146. Increased rates of conjugation can result from the antibiotic-mediated induction of the bacterial SOS response and increased membrane permeability153–157, which may stem from the DNA damage inflicted by certain antibiotics. However, the effects of antibiotics on the rates of HGT (particularly conjugation) are contested, with other studies demonstrating highest plasmid transfer rates in the absence of antibiotics158–161. These conflicting reports are likely to arise from the difficulty of disentangling the effects of antibiotics on HGT rates from the antibiotic-mediated selection of transconjugants (Fig. 3), which suggests that the reported post-antibiotic enrichments of ARG-encoding MGEs often result from the growth advantage conferred on transconjugants, not higher HGT rates per se162.
Box 2. Mechanisms of horizontal gene transfer in the gut microbiome.
Transformation involves bacterial uptake of extracellular DNA (see the figure, part a). During transduction, bacteriophages serve as vectors for the horizontal transfer of genetic material, including antimicrobial resistance genes, between bacterial hosts (see the figure, part b). Conjugation describes the direct transfer of genetic material from a bacterial donor to a bacterial recipient, a process that necessitates physical contact between the two cells (see the figure, part c).
Fig. 3|. Assessing the effect of antibiotics on horizontal gene transfer rates.

a, A community in equilibrium is depicted. In the absence of an antibiotic selection pressure, the basal horizontal gene transfer rates are maintained. The chart on the right depicts transconjugants as a fraction of the total cells in the community. b,c, During antibiotic treatment, either conjugation rates are increased (part b) or selection promotes the growth of plasmid-containing populations (part c); these two outcomes could also take place concurrently. Both cases result in a higher fraction of transconjugants relative to the community in the absence of antibiotics. This leads to difficulties in distinguishing the effects of antibiotics on conjugation rates from those of antibiotic-mediated selection.
Investigation of HGT rates within the gut microbiome is still in its nascency; our understanding of how the frequency of horizontal transfer is affected by antibiotics is very limited, let alone our grasp on the variability of this impact across drug classes. Further investigation into the effects of antibiotics on HGT are needed, with careful experimental design that enables the precise determination of the frequency of HGT while controlling for the effects of antibiotics on other aspects of bacterial biology (such as the growth rate). Furthermore, although shotgun sequencing has enabled a comprehensive characterization of the taxonomic and resistance profiles of microbial communities (albeit with limitations noted earlier), technological limitations of short-read sequencing make it ill-suited for investigations of HGT (Box 3). For one, short-read metagenome assemblies are highly fragmented and cannot be used for identification of genetic origins (that is, whether from chromosome or plasmid) and taxonomic origins of ARGs151. Hybrid assemblies (based on both short and long reads) can address many of these limitations, enabling the contextualized characterization of ARGs28. Additionally, because of the total DNA extraction step that typically precedes sequencing library preparation, shotgun metagenomic sequencing does not enable the efficient linking of MGEs with bacterial hosts, obscuring community HGT dynamics. These limitations can be addressed through Hi-C, a technique in which crosslinking before cell lysis preserves interacting DNA molecules and the information regarding the MGE–bacterial host associations138. Hence, in addition to the aforementioned controls, future investigations into HGT dynamics would benefit from application of long-read sequencing and Hi-C platforms, offering further insight into the frequency and direction of ARG mobilization within the context of an antibiotic-perturbed microbiome.
Box 3. Approaches for profiling the mobile fraction of the community resistome.
To perform shotgun sequencing, the total metagenomic DNA of the community is first extracted and then sequenced to produce short (~150–300 bp) sequencing reads149. The reads can subsequently be assembled into larger contigs, which are then grouped into bins based on the predicted common microbial origin149 (see the figure). Despite computation advancements in assembly, binning and classification, many of the assembled contigs, particularly those corresponding to mobile genetic elements (MGEs), remain unbinned, obscuring their taxonomic origins150. Furthermore, the relatively short length of the assembled contigs makes it impractical to determine the genomic origins (chromosome versus plasmid) of these contigs151. Consequently, although shotgun metagenomics enables a comprehensive characterization of antibiotic resistance genes (ARGs), the information regarding the taxonomic and genomic origins of ARGs is lost. Recent developments in long-read sequencing can address many of these limitations. Longer reads often enable the assembly of complete (that is, circular) plasmidic contigs and yield much more informative (if not complete) chromosomal contigs152. This increases the efficiency of identification of the genomic origins of ARGs and pairing of chromosomal ARGs to taxa of origin. The longer contigs also reveal the genetic context of ARGs and their potential localization within MGEs151. However, it should be noted that the sequencing error rates of long-read sequencing platforms are often substantially higher than those of short-read methods; hence, a hybrid approach, taking advantage of longer contigs of long-read sequencing and higher accuracy of short-read sequencing, presents an optimal approach151. Last, given that the total metagenomic DNA is similarly extracted before sequencing, the information regarding the MGE–chromosome associations is lost. To this end, crosslinking of interacting DNAs before cellular lysis preserves the chromosome–MGE pairs. Hi-C, in which this strategy is used, yields hybrid contigs that can be used to identify the MGE hosts138.
Stars represent the location of ARGs.
Towards predictive models
The growing body of research directed at elucidating antibiotic-induced microbiome perturbations could ultimately grant us the capacity to model the post-treatment microbiome dynamics and to forecast off-target therapeutic effects, guiding the selection of treatments that minimize collateral microbiome damage. However, the immense complexity and interindividual variation of the gut microbiome is well established, with its composition affected by innumerable factors. Consequently, and as we describe in previous sections, the post-antibiotic community dynamics depends on (and is confounded by) numerous treatment, microbial, host and environmental factors (Table 1), making it currently non-trivial to generate accurate generalized predictive models of microbiome or host outcomes related to antibiotic exposure. Nonetheless, some early efforts in generating predictive models of microbiome perturbation in specific cases and populations have been made by accounting for and, when possible, controlling for the variables of interest (Table 1). For instance, in a study involving a cohort of preterm infants from a single hospital, and testing over 70 metadata variables, a model based on only six predictors (postmenstrual age, breast milk consumption, CRIB (Clinical Risk Index for Babies) II score, and administration of meropenem, cefotaxime and ticarcillin–clavulanate) explained 33% of the variance in the species richness in this cohort52. Another investigation involving infants within the same hospital system similarly found that a predictive model based on day of life (age) and antibiotic treatment information alone could account for 57% of the variability in Shannon diversity126. Thus, despite the complexity of the compositional dynamics of the microbiome, control over deterministic factors through careful selection of the study cohort can enable the development of accurate predictive models in specific settings. Conversely, careful selection of variables to control often decreases the generalizability of the study (for example, successful prediction of microbiome outcomes may be confined to only preterm infants in the region or hospital under investigation). To generate models with higher predictive accuracy and breadth, it is important that future investigations into the compositional consequences of microbiome perturbation integrate extensive metadata on treatment, microbial and host variables, some of which are outlined in Table 1. Moreover, it is imperative to subject the resulting models to cross-cohort validation to determine the generalizability of the underlying predictors.
Ameliorating perturbations
The gastrointestinal tract requires a high, diverse microbial load to carry out requisite microbiome functions163. Thus, efforts towards the maintenance or restoration of richness and diversity in the gut microbiome on antibiotic exposure are of extreme medical importance113,164,165. Here, we present an overview of approaches (established and in development) aimed at either minimizing antibiotic-induced damage to the gut community during treatment or restoring microbiome diversity post-exposure (Fig. 4). Furthermore, alternative strategies with improved precision and decreased off-target effects relative to antibiotics are urgently needed and could be used to mitigate treatment-induced gut dysbiosis. Engineered probiotic microorganisms have shown great promise in treatment and prevention of pathogenic colonization with increasing precision, and may offer an avenue for the development of the aforementioned alternative anti-infective strategies166. Given this potential, we discuss the use of engineered probiotic therapies with demonstrated anti-pathogen activity167,168. Based on the shortcomings of many of these approaches, or the likelihood of community evolution to outpace a given therapy, we expect that the most successful therapies in the future are likely to come from a combination of approaches in mitigating gut dysbiosis and preventing gastrointestinal infections.
Fig. 4|. Approaches aimed at maintaining or restoring gut microbiome structure on antibiotic treatment.

Antibiotics reduce the microbial richness and diversity of the gut microbiome. The gut community post-antibiotics can be restored via community replacement (faecal microbiota transplant (FMT)) or prebiotic administration, which may facilitate the growth of commensal members of the microbiome and/or prevent the colonization of pathogenic bacteria. Alternatively, antibiotic-mediated destruction of the microbiome can be prevented by bioengineering approaches targeting the antibiotic. Specifically, the gut concentrations of the drugs can be diminished through enzymatic degradation or physical sequestration of antibiotics, preventing a broader reduction in the richness and diversity of the gut community. Last, the narrowing of the spectrum of activity of the treatment regimen might also prevent broader damage to the gut microbial community. This could be achieved by choosing antibiotics with inherently narrower activity spectra or through combinations of antibiotics with other drugs, which may similarly result in a narrower spectrum of antimicrobial activity.
Minimizing antibiotic-induced perturbations to the gut
Given that antibiotics vary in their spectrum of activity against the diverse members of the commensal gut microbiome, the choice of treatments with a narrower spectrum could result in less perturbation to the microbiome and faster community recovery169. For instance, relative to vancomycin treatment (broad spectrum), fidaxomicin treatment (narrow spectrum) resulted in a smaller shift from the pretreatment taxonomic structure of the mouse gut, promoted a faster recovery of the taxonomic structure and preserved the C. difficile colonization resistance capacity of the gut to a higher extent169. However, it should be noted that the in vitro spectra of activities do not directly translate in vivo, and, as mentioned previously, higher levels of gut microbiome perturbation may result from agents with narrower in vitro spectra. The specificity of antibiotics can also be modulated through coadministration of other antibiotic and non-antibiotic medications: combination treatments can have differential effects (neutral, synergistic or antagonistic) on the activity of the given antibiotic against microorganisms, and this modulation is variable across microbial taxa170. Correspondingly, it may be possible to achieve species-specific treatment by using drug combinations that have synergistic effects against pathogens and antagonistic effects against commensal microorganisms113,170.
During systemic administration of antibiotics (for example, in response to suspected bloodstream infections), reducing the levels of antimicrobials in the gut can also minimize off-target treatment effects and damage to the gut microbiome. As proof of principle, gut commensal Bacteroides spp. can release cephalosporinases that protect susceptible members of the community from lethal doses of cefotaxime171. Similarly, oral administration of a β-lactamase concurrently with systemic β-lactam antibiotic treatment maintains the taxonomic structure of the gut community164,172. Although coadministration of the β-lactamase reduced the antibiotic concentrations in the gut, the serum levels of antimicrobials remained unaltered164. Production and purification of antibiotic-inactivating agents at scale is non-trivial; expression and delivery of these agents by engineered probiotics provides a viable alternative. The probiotic Lactococcus lactis was engineered to express and secrete the TEM1 β-lactamase; oral gavage of the engineered microorganism with concomitant intraperitoneal injections of ampicillin reduced the gut levels of the drug (without affecting the serum concentrations of the antibiotic), lessened collateral damage to the gut microbiome and maintained C. difficile colonization resistance in mice173. Notably, TEM1 was designed to be expressed by two gene segments as two enzymatically inactive components that reconstitute the functional enzyme on secretion: this split nature of the gene minimizes the risk of its spread through HGT173.
Antibiotic adsorbing agents have also been used in the context of the gut microbiome, as they have the advantage of inactivating a broad range of compounds. The most notable example is DAV132, an adsorbent based on activated charcoal, which reduces the faecal concentrations of moxifloxacin and fluoroquinolones without altering the serum levels of the antibiotics165,174. DAV132 can further adsorb antibiotics belonging to five other drug classes ex vivo165. Coadministration of DAV132 with antibiotics protects gut bacterial richness and diversity, impedes the spread of pathobionts and resistant organisms, and maintains the colonization resistance capacity of the gut against C. difficile165,174,175. DAV132 can be further modified to achieve site-specific inactivation of antibiotics as well: coating of the adsorbent in a pH-dependent polymer ensured its release and activity in the colon, without compromising the antibiotic activity in the small intestine176.
Prebiotic-mediated expansion of beneficial taxa
Given the metabolic capacity of the microbiome to digest complex dietary fibres, prebiotic compounds (which are compounds that stimulate the growth of beneficial organisms in the microbiome) have been increasingly used to both combat enteropathogens and restore commensal gut microbiome structure during or post-perturbation. Antibiotics disrupt the production of SCFAs in the gut. Polysaccharide-rich diets can be used to restore the production of these molecules; this restoration is largely associated with colonization resistance against pathogens49,177,178. Defined polysaccharides have been examined in the context of mitigating C. difficile infection. In one study, purified mucin glycans were used to increase community diversity post-antibiotic treatment; this activity decreases the proliferation of C. difficile on infection179. In another study, dietary xanthan gum demonstrated a capacity to increase fibre-degrading taxa, although without significantly altering alpha diversity, and prevented C. difficile colonization, probably owing to the maintenance of the commensal microbiota during antibiotic treatment180. In addition to restructuring the microbiome, it is also possible that several soluble non-starch polysaccharides could serve as a physical barrier against pathogen adherence to the epithelial barrier181,182. Most recently, a symbiotic combination, Bifidobacterium infantis and milk oligosaccharides, was used to stably reverse dysbiotic human microbiomes183.
Restoring antibiotic-perturbed gut communities via faecal microbiota transplants
The microbiome structure can be restoredvia whole-community replacement, or faecal microbiota transplants (FMTs), from healthy donors. Originally designed for its use in the treatment of recurrent C. difficile infection (rCDI), the use of FMTs has expanded to other infectious diseases, including the treatment of enteric infections, MDR bacterial colonization and viral infections (as reviewed elsewhere)184. Following antibiotic treatment, administration of an autologous FMT resulted in faster restoration of the microbiome than a 12-member probiotic cocktail, which notably delayed the recovery in taxonomic diversity in the gut microbiome185. In the context of rCDI, several microbial communities (derived from human donor stool or from defined bacterial culture) have shown promise in providing decreased risk of recurrent episodes186–189. The most recent success story is SER-109 (produced by Seres Therapeutics), a compilation of purified Firmicutes spores that shows success against rCDI in phase III clinical trials188. Importantly, SER-109 improves the production of secondary bile acids, which presumably prevent C. difficile germination190,191. Another defined community, VE303 (produced by Vedanta Biosciences), is composed of fewer members than SER-109 and demonstrates robust restoration of butyrate production and secondary bile acid production in dysbiotic human microbiomes engrafted into mice; this community is well tolerated when administered with vancomycin to healthy volunteers189. In all, FMTs and defined communities have been generally demonstrated to be safe and hold great promise in post-antibiotic restoration of the gut community. Nonetheless, there are noteworthy therapeutic risks, including the unintended introduction of pathogens and ARGs into the gut of the recipient185,192. In the case of the microbiome restoration product RBX2660, although there is a clear benefit against rCDI, high-resolution profiling of microbiome dynamics indicated that it also engrafted donor antibiotic-resistant organisms and ARGs into the new host192. These unintended consequences may prove dangerous to patients; although rarely, previous FMT administration has resulted in severe opportunistic infections from a donor pathobiont193,194. As therapies such as SER-109 and VE303 are further evaluated, it will be important to identify recipient microbiome-specific effects on community trajectory post-administration.
Probiotics as alternatives to antibiotics
Probiotics, which are live microorganisms with potential health benefits195, can provide alternatives to antibiotics in treatment and prevention of microbial infections. However, probiotics may perform less well than whole-community replacement strategies (discussed above) in microbiome restoration and could result in resistome expansion in an antibiotic-pretreated gut196. Nonetheless, probiotic organisms have been extensively used therapeutically against various pathogens, commonly achieving their native anti-infective activity through competitive exclusion, sequestration and release of antimicrobial compounds (constitutive or induced), as reviewed elsewhere166. Recent expansions of the toolsets aimed at genetic modification of bacterial and yeast probiotics197–199 have enabled the engineering of probiotic organisms with more potent and/or targeted anti-pathogenic activities. For instance, probiotic Lactobacillus casei strains were engineered to express and display adhesion proteins that enabled them to robustly colonize the intestine; the engineered probiotic prevented the intestinal colonization by a foodborne pathogen, Listeria monocytogenes, and protected mice from lethal infection167. Similarly, the anti-pathogen activity of the probiotic Escherichia coli Nissle 1917 was enhanced by expressing nanobodies as bacterial curli fibre fusions, which enables the engineered strains to sequester pathogens (for example, enteropathogenic E. coli and Shigella flexneri) or pathogen toxins (for example, Shiga toxin and C. difficile toxin TcdA)200.
Probiotics can also be engineered to express and release pathogen-specific antimicrobial agents201. To further reduce the off-target effects of the treatment, the release of antimicrobials can be engineered to be induced on sensing of external stimuli produced by the target pathogen202,203. For example, E. coli Nissle 1917 was engineered to lyse itself on sensing the Pseudomonas aeruginosa quorum-sensing molecules, releasing cytosolic lysis and anti-biofilm molecules204. The engineered probiotic reduced gut P. aeruginosa titres in both mouse and Caenorhabditis elegans infection models204. The release of antimicrobials can alternatively be engineered to be induced by molecules characteristic of gut damage (dysbiosis and inflammation)205. Notably, probiotic E. coli was engineered to sense extracellular sialic acid, a proxy for gut dysbiosis, and respond by producing a bile salt hydrolase, deconjugating primary bile salts in the gut168. The engineered probiotic reduced the germination of C. difficile endospores, resulting in 100% survival and improved clinical symptoms in murine models of C. difficile infection168.
Conclusion and future directions
Because the gut microbiome is increasingly linked to human health, research on how antibiotics reshape the microbiome and endanger the health of the host is of considerable interest for public health. Furthermore, a greater understanding of the mechanisms by which antibiotics perturb the human microbiome is critical to maximizing the therapeutic benefits of these agents while reducing both the damage imposed on the commensal gut microbiome and the broader spread of antimicrobial resistant pathogens23,24. As we have outlined here, the gut microbiome response to antibiotics is highly complex and variable33, driven by a multitude of factors33,59,206,207. To quantitatively understand and predict the most important biological variables that influence antibiotic-mediated destruction of this complex ecology, techniques for host-informed sample collection168, sequencing methodologies and statistical analyses need to be standardized within the field. Recent collaborative advocacy for standardized reporting guidelines for microbiome studies will enable the field to move towards this goal208.
In the effort to achieve globally robust predictions of antibiotic–microbiome outcomes, there lies a difficulty in balancing generalizability with reproducibility. Specifically, it is imperative for future models of antibiotic-mediated damage to the gut community to control for and test the predictive capacities of broader sets of host and microbial variables to yield more predictive models of the community dynamics52,59,126. Among the notable variables that are traditionally overlooked in microbiome studies is geographical origin of the studied population. Investigations of the gut microbiome have predominantly focused on industrialized populations in the USA and Europe, representing a minority of the world population. Thus, our current models of microbiome dynamics are critically limited in their scope and application209.
The proliferation of high-throughput, low-cost sequencing technologies in the academic, medical and private sectors provides the field with an unprecedented opportunity to further develop predictive statistical methods that can be used as part of a public health framework to prevent antibiotic resistance emergence and future infections. Complementing such strategies, a wealth of scholarship highlights the translational potential of emerging microbiome restorative strategies, such as narrow-spectrum engineered probiotics167,168,173,200,204,205 or defined FMTs185–188,190–192. To realize the frequently touted goal of ‘personalized medicine’ in the treatment of infectious diseases, much basic and translational work is still needed to accurately understand and predict the host, microbial and environmental factors that control the response of the microbiome to specific antimicrobials, the restoration of pre-perturbation microbiome composition and function, and the response to novel microbiome-directed therapeutics.
Supplementary Material
Acknowledgements
Work in the authors’ laboratory was supported in part by awards to G.D. through the National Institute of Allergy and Infectious Diseases (NIAID; award numbers U01AI123394 and R01AI155893), the Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD; award number R01HD092414), and the National Center for Complementary and Integrative Health (NCCIH; award number R01AT009741) of the National Institutes of Health (NIH); through the National Institute for Occupational Safety and Health (NIOSH) of the US Centers for Disease Control and Prevention (CDC; award number R01OH011578); and through the Agency for Healthcare Research and Quality (AHRQ) of the US Department of Health & Human Services (DHHS; award number R01HS027621). S.R.S.F. was supported by an award from Paediatric Gastroenterology Research Training Program (T32DK077653) through the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding agencies.
Footnotes
Peer review information Nature Reviews Microbiology thanks Ilana Brito and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Competing interests
The authors declare no competing interests.
Additional information
Supplementary information The online version contains supplementary material available at https://doi.org/10.1038/s41579-023-00933-y.
References
- 1.Gomaa EZ Human gut microbiota/microbiome in health and diseases: a review. Antonie van Leeuwenhoek 113, 2019–2040 (2020). [DOI] [PubMed] [Google Scholar]
- 2.Derrien M, Alvarez AS & de Vos WM The gut microbiota in the first decade of life. Trends Microbiol 27, 997–1010 (2019). [DOI] [PubMed] [Google Scholar]
- 3.Gilbert JA et al. Current understanding of the human microbiome. Nat. Med 24, 392–400 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Backhed F, Ley RE, Sonnenburg JL, Peterson DA & Gordon JI Host–bacterial mutualism in the human intestine. Science 307, 1915–1920 (2005). [DOI] [PubMed] [Google Scholar]
- 5.Nguyen LH et al. Antibiotic use and the development of inflammatory bowel disease: a national case-control study in Sweden. Lancet Gastroenterol. Hepatol 5, 986–995 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cox LM & Blaser MJ Antibiotics in early life and obesity. Nat. Rev. Endocrinol 11, 182–190 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Farhana L, Banerjee HN, Verma M & Majumdar APN Role of microbiome in carcinogenesis process and epigenetic regulation of colorectal cancer. Methods Mol. Biol 1856, 35–55 (2018). [DOI] [PubMed] [Google Scholar]
- 8.Akimoto N et al. Rising incidence of early-onset colorectal cancer — a call to action. Nat. Rev. Clin. Oncol 18, 230–243 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Johnson S et al. Epidemics of diarrhea caused by a clindamycin-resistant strain of Clostridium difficile in four hospitals. N. Engl. J. Med 341, 1645–1651 (1999). [DOI] [PubMed] [Google Scholar]
- 10.Kim JH et al. Maternal antibiotic exposure during pregnancy is a risk factor for community-acquired urinary tract infection caused by extended-spectrum beta-lactamase-producing bacteria in infants. Pediatr. Nephrol 37, 163–170 (2022). [DOI] [PubMed] [Google Scholar]
- 11.Lynch IJ & Martinez FJ Clinical relevance of macrolide-resistant Streptococcus pneumoniae for community-acquired pneumonia. Clin. Infect. Dis 34, S27–S46 (2002). [DOI] [PubMed] [Google Scholar]
- 12.Fleming A On the antibacterial action of cultures of a penicillium, with special reference to their use in the isolation of B. influenzae. 1929. Bull. World Health Organ 79, 780–790 (2001). [PMC free article] [PubMed] [Google Scholar]
- 13.Adedeji WA The treasure called antibiotics. Ann. Ib. Postgrad. Med 14, 56–57 (2016). [PMC free article] [PubMed] [Google Scholar]
- 14.Centers for Disease Control and Prevention. Life Expectancy. Centers for Disease Control and Prevention https://www.cdc.gov/nchs/fastats/life-expectancy.htm (2021).
- 15.Ng KM et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature 502, 96–99 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Fishbein SRS et al. Randomized controlled trial of oral vancomycin treatment in Clostridioides difficile-colonized patients. mSphere doi: 10.1128/mSphere.00936-20 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Centers for Disease Control and Prevention. Antibiotic resistance threats in the United States. Centers for Disease Control and Prevention; https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf (2019). [Google Scholar]
- 18.Anthony WE, Burnham CD, Dantas G & Kwon JH The gut microbiome as a reservoir for antimicrobial resistance. J. Infect. Dis 223, S209–S213 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Hayase E et al. Mucus-degrading Bacteroides link carbapenems to aggravated graft-versus-host disease. Cell 185, 3705–3719.e14 (2022). This study highlights an underexplored area in identifying antibiotic-induced microbiome perturbation as a contributor to intestinal graft-versus-host disease.
- 20.Shono Y et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl Med 8, 339ra371 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wypych TP & Marsland BJ Antibiotics as instigators of microbial dysbiosis: implications for asthma and allergy. Trends Immunol 39, 697–711 (2018). [DOI] [PubMed] [Google Scholar]
- 22.Klein EY et al. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc. Natl Acad. Sci. USA 115, E3463–E3470 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gould IM A review of the role of antibiotic policies in the control of antibiotic resistance. J. Antimicrob. Chemother 43, 459–465 (1999). [DOI] [PubMed] [Google Scholar]
- 24.Olesen SW et al. The distribution of antibiotic use and its association with antibiotic resistance. eLife doi: 10.7554/eLife.39435 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 399, 629–655 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Sommer MOA, Dantas G & Church GM Functional characterization of the antibiotic resistance reservoir in the human microflora. Science 325, 1128–1131 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gasparrini AJ et al. Antibiotic perturbation of the preterm infant gut microbiome and resistome. Gut Microbes 7, 443–449 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mahmud B et al. Epidemiology of plasmid lineages mediating the spread of extended-spectrum beta-lactamases among clinical Escherichia coli. mSystems doi: 10.1128/msystems.00519-22 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Forster SC et al. Strain-level characterization of broad host range mobile genetic elements transferring antibiotic resistance from the human microbiome. Nat. Commun 13, 1445 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Stracy M et al. Minimizing treatment-induced emergence of antibiotic resistance in bacterial infections. Science 375, 889–894 (2022). Using a machine-learning approach, this study leverages a massive bacterial genomics database tied to infection data to identify genomic predictors by which treatment-induced emergence of antibiotic resistance can be avoided.
- 31.Dethlefsen L, Huse S, Sogin ML & Relman DA The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol 6, e280 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Palleja A et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat. Microbiol 3, 1255–1265 (2018). [DOI] [PubMed] [Google Scholar]
- 33.Anthony WE et al. Acute and persistent effects of commonly used antibiotics on the gut microbiome and resistome in healthy adults. Cell Rep 39, 110649 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Dubinsky V et al. Predominantly antibiotic-resistant intestinal microbiome persists in patients with pouchitis who respond to antibiotic therapy. Gastroenterology 158, 610–624.e13 (2020). [DOI] [PubMed] [Google Scholar]
- 35.Van Engelen TSR et al. Gut microbiome modulation by antibiotics in adult asthma: a human proof-of-concept intervention trial. Clin. Gastroenterol. Hepatol 20, 1404–1407.e4 (2022). [DOI] [PubMed] [Google Scholar]
- 36.Doan T et al. Mass azithromycin distribution and community microbiome: a cluster-randomized trial. Open Forum Infect. Dis 5, ofy182 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Reyman M et al. Effects of early-life antibiotics on the developing infant gut microbiome and resistome: a randomized trial. Nat. Commun 13, 893 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Raymond F et al. The initial state of the human gut microbiome determines its reshaping by antibiotics. ISME J 10, 707–720 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Reijnders D et al. Short-term microbiota manipulation and forearm substrate metabolism in obese men: a randomized, double-blind, placebo-controlled trial. Obes. Facts 11, 318–326 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zarrinpar A et al. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat. Commun 9, 2872 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kelly CP et al. Saccharomyces boulardii CNCM I-745 modulates the fecal bile acids metabolism during antimicrobial therapy in healthy volunteers. Front. Microbiol 10, 336 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Tsukuda N et al. Key bacterial taxa and metabolic pathways affecting gut short-chain fatty acid profiles in early life. ISME J 15, 2574–2590 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Young VB & Schmidt TM Antibiotic-associated diarrhea accompanied by large-scale alterations in the composition of the fecal microbiota. J. Clin. Microbiol 42, 1203–1206 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lloyd-Price J et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Theriot CM et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun 5, 3114 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Isaac S et al. Microbiome-mediated fructose depletion restricts murine gut colonization by vancomycin-resistant Enterococcus. Nat. Commun 13, 7718 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Giel JL, Sorg JA, Sonenshein AL & Zhu J Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PLoS ONE 5, e8740 (2010). This study underscores the importance of microbial transformation of bile salts in conferring microbiome-mediated colonization resistance to a spore-forming pathogen.
- 48.Reed AD, Nethery MA, Stewart A, Barrangou R & Theriot CM Strain-dependent inhibition of Clostridioides difficile by commensal clostridia carrying the bile acid-inducible (bai) operon. J. Bacteriol doi: 10.1128/JB.00039-20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Guinan J, Wang S, Hazbun TR, Yadav H & Thangamani S Antibiotic-induced decreases in the levels of microbial-derived short-chain fatty acids correlate with increased gastrointestinal colonization of Candida albicans. Sci. Rep 9, 8872 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Buffie CG & Pamer EG Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol 13, 790–801 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.MacPherson CW et al. Gut bacterial microbiota and its resistome rapidly recover to basal state levels after short-term amoxicillin-clavulanic acid treatment in healthy adults. Sci. Rep 8, 11192 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Gibson MK et al. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat. Microbiol 1, 16024 (2016). This study shows that in infant guts, the ARGs enriched after antibiotic treatment highly correlate with the abundance of single species. This study also utilizes functional screening to identify novel ARGs in stool metagenomes.
- 53.Loo VG et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile-associated diarrhea with high morbidity and mortality. N. Engl. J. Med 353, 2442–2449 (2005). [DOI] [PubMed] [Google Scholar]
- 54.Weill FX et al. Genomic insights into the 2016–2017 cholera epidemic in Yemen. Nature 565, 230–233 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Reijnders D et al. Effects of gut microbiota manipulation by antibiotics on host metabolism in obese humans: A randomized double-blind placebo-controlled trial. Cell Metab 24, 63–74 (2016). [DOI] [PubMed] [Google Scholar]
- 56.Gao H et al. Antibiotic exposure has sex-dependent effects on the gut microbiota and metabolism of short-chain fatty acids and amino acids in mice. mSystems doi: 10.1128/mSystems.00048-19 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sim CK et al. A mouse model of occult intestinal colonization demonstrating antibiotic-induced outgrowth of carbapenem-resistant Enterobacteriaceae. Microbiome 10, 43 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Stewardson AJ et al. Collateral damage from oral ciprofloxacin versus nitrofurantoin in outpatients with urinary tract infections: a culture-free analysis of gut microbiota. Clin. Microbiol. Infect 21, 344.e1–344.e11 (2015). [DOI] [PubMed] [Google Scholar]
- 59.Willmann M et al. Distinct impact of antibiotics on the gut microbiome and resistome: a longitudinal multicenter cohort study. BMC Biol 17, 76 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hinterwirth A et al. Rapid reduction of Campylobacter species in the gut microbiome of preschool children after oral azithromycin: a randomized controlled trial. Am. J. Trop. Med. Hyg 103, 1266–1269 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chaima D et al. Biannual administrations of azithromycin and the gastrointestinal microbiome of Malawian children: a nested cohort study within a randomized controlled trial. Front. Public Health 10, 756318 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Arzika AM et al. Gut resistome of preschool children after prolonged mass azithromycin distribution: a cluster-randomized trial. Clin. Infect. Dis 73, 1292–1295 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Doan T et al. Macrolide and nonmacrolide resistance with mass azithromycin distribution. N. Engl. J. Med 383, 1941–1950 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Doan T et al. Macrolide resistance in MORDOR I — a cluster-randomized trial in Niger. N. Engl. J. Med 380, 2271–2273 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Doan T et al. Gut microbiome alteration in MORDOR I: a community-randomized trial of mass azithromycin distribution. Nat. Med 25, 1370–1376 (2019). [DOI] [PubMed] [Google Scholar]
- 66.Pickering H et al. Impact of azithromycin mass drug administration on the antibiotic-resistant gut microbiome in children: a randomized, controlled trial. Gut Pathog 14, 5 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Oldenburg CE et al. Effect of commonly used pediatric antibiotics on gut microbial diversity in preschool children in Burkina Faso: a randomized clinical trial. Open Forum Infect. Dis 5, ofy289 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yassour M et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl Med 8, 343ra381 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zlitni S et al. Strain-resolved microbiome sequencing reveals mobile elements that drive bacterial competition on a clinical timescale. Genome Med 12, 50 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Goltsman DSA et al. Metagenomic analysis with strain-level resolution reveals fine-scale variation in the human pregnancy microbiome. Genome Res 28, 1467–1480 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Olm MR et al. Necrotizing enterocolitis is preceded by increased gut bacterial replication, Klebsiella, and fimbriae-encoding bacteria. Sci. Adv 5, eaax5727 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Brito IL et al. Transmission of human-associated microbiota along family and social networks. Nat. Microbiol 4, 964–971 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Vinarov Z et al. Impact of gastrointestinal tract variability on oral drug absorption and pharmacokinetics: An UNGAP review. Eur. J. Pharm. Sci 162, 105812 (2021). [DOI] [PubMed] [Google Scholar]
- 74.Rao S, Kupfer Y, Pagala M, Chapnick E & Tessler S Systemic absorption of oral vancomycin in patients with Clostridium difficile infection. Scand. J. Infect. Dis 43, 386–388 (2011). [DOI] [PubMed] [Google Scholar]
- 75.Connelly S, Subramanian P, Hasan NA, Colwell RR & Kaleko M Distinct consequences of amoxicillin and ertapenem exposure in the porcine gut microbiome. Anaerobe 53, 82–93 (2018). [DOI] [PubMed] [Google Scholar]
- 76.Singh J, Burr B, Stringham D & Arrieta A Commonly used antibacterial and antifungal agents for hospitalised paediatric patients: implications for therapy with an emphasis on clinical pharmacokinetics. Paediatr. Drugs 3, 733–761 (2001). [DOI] [PubMed] [Google Scholar]
- 77.Leopold SR et al. Murine model for measuring effects of humanized-dosing of antibiotics on the gut microbiome. Front. Microbiol 13, 813849 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Browne HP et al. Host adaptation in gut Firmicutes is associated with sporulation loss and altered transmission cycle. Genome Biol 22, 204 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fawley WN et al. Efficacy of hospital cleaning agents and germicides against epidemic Clostridium difficile strains. Infect. Control. Hosp. Epidemiol 28, 920–925 (2007). [DOI] [PubMed] [Google Scholar]
- 80.Baines SD, O’Connor R, Saxton K, Freeman J & Wilcox MH Activity of vancomycin against epidemic Clostridium difficile strains in a human gut model. J. Antimicrob. Chemother 63, 520–525 (2009). [DOI] [PubMed] [Google Scholar]
- 81.Antunes A et al. Global transcriptional control by glucose and carbon regulator CcpA in Clostridium difficile. Nucleic Acids Res 40, 10701–10718 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Normington C et al. Biofilms harbour Clostridioides difficile, serving as a reservoir for recurrent infection. NPJ Biofilms Microbiomes 7, 16 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Theriot CM, Bowman AA & Young VB Antibiotic-induced alterations of the gut microbiota alter secondary bile acid production and allow for Clostridium difficile spore germination and outgrowth in the large intestine. mSphere doi: 10.1128/mSphere.00045-15 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Duncan K, Carey-Ewend K & Vaishnava S Spatial analysis of gut microbiome reveals a distinct ecological niche associated with the mucus layer. Gut Microbes 13, 1874815 (2021). This analysis utilizes a number of imaging techniques to characterize the microbial composition of the mucus layer in a mouse.
- 85.Elhenawy W et al. High-throughput fitness screening and transcriptomics identify a role for a type IV secretion system in the pathogenesis of Crohn’s disease-associated Escherichia coli. Nat. Commun 12, 2032 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bakkeren E et al. Pathogen invasion-dependent tissue reservoirs and plasmid-encoded antibiotic degradation boost plasmid spread in the gut. eLife doi: 10.7554/eLife.69744 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bakkeren E et al. Salmonella persisters promote the spread of antibiotic resistance plasmids in the gut. Nature 573, 276–280 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Cabral DJ et al. Microbial metabolism modulates antibiotic susceptibility within the murine gut microbiome. Cell Metab 30, 800–823.e7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chambers SA et al. A solution to antifolate resistance in group B Streptococcus: untargeted metabolomics identifies human milk oligosaccharide-induced perturbations that result in potentiation of trimethoprim. mBio doi: 10.1128/mBio.00076-20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Liu Y et al. Gut microbiome alterations in high-fat-diet-fed mice are associated with antibiotic tolerance. Nat. Microbiol 6, 874–884 (2021). [DOI] [PubMed] [Google Scholar]
- 91.Vega NM, Allison KR, Samuels AN, Klempner MS & Collins JJ Salmonella typhimurium intercepts Escherichia coli signaling to enhance antibiotic tolerance. Proc. Natl Acad. Sci. USA 110, 14420–14425 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zarkan A et al. Inhibition of indole production increases the activity of quinolone antibiotics against E. coli persisters. Sci. Rep 10, 11742 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Varga JJ et al. Antibiotics drive expansion of rare pathogens in a chronic infection microbiome model. mSphere 7, e0031822 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Amor DR & Gore J Fast growth can counteract antibiotic susceptibility in shaping microbial community resilience to antibiotics. Proc. Natl Acad. Sci. USA 119, e2116954119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Bottery MJ et al. Inter-species interactions alter antibiotic efficacy in bacterial communities. ISME J 16, 812–821 (2022). This study utilizes a simple defined community framework to identify quantitative features of a small bacterial community that predict antibiotic susceptibility in the community.
- 96.Ng KM et al. Recovery of the gut microbiota after antibiotics depends on host diet, community context, and environmental reservoirs. Cell Host Microbe 26, 650–665.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Isaac S et al. Short- and long-term effects of oral vancomycin on the human intestinal microbiota. J. Antimicrob. Chemother 72, 128–136 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Coyte KZ & Rakoff-Nahoum S Understanding competition and cooperation within the mammalian gut microbiome. Curr. Biol 29, R538–R544 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Aranda-Diaz A et al. Bacterial interspecies interactions modulate pH-mediated antibiotic tolerance. eLife doi: 10.7554/eLife.51493 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Adamowicz EM, Flynn J, Hunter RC & Harcombe WR Cross-feeding modulates antibiotic tolerance in bacterial communities. ISME J 12, 2723–2735 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Adamowicz EM, Muza M, Chacon JM & Harcombe WR Cross-feeding modulates the rate and mechanism of antibiotic resistance evolution in a model microbial community of Escherichia coli and Salmonella enterica. PLoS Pathog 16, e1008700 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hazleton KZ et al. Dietary fat promotes antibiotic-induced Clostridioides difficile mortality in mice. NPJ Biofilms Microbiomes 8, 15 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Guasch-Ferre M et al. Dietary fat intake and risk of cardiovascular disease and all-cause mortality in a population at high risk of cardiovascular disease. Am. J. Clin. Nutr 102, 1563–1573 (2015). [DOI] [PubMed] [Google Scholar]
- 104.Leone V, Chang EB & Devkota S Diet, microbes, and host genetics: the perfect storm in inflammatory bowel diseases. J. Gastroenterol 48, 315–321 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Lee JY et al. High-fat diet and antibiotics cooperatively impair mitochondrial bioenergetics to trigger dysbiosis that exacerbates pre-inflammatory bowel disease. Cell Host Microbe 28, 273–284.e6 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cabral DJ, Wurster JI, Korry BJ, Penumutchu S & Belenky P Consumption of a Western-style diet modulates the response of the murine gut microbiome to ciprofloxacin. mSystems doi: 10.1128/mSystems.00317-20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wurster JI et al. Streptozotocin-induced hyperglycemia alters the cecal metabolome and exacerbates antibiotic-induced dysbiosis. Cell Rep. 37, 110113 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Tanes C et al. Role of dietary fiber in the recovery of the human gut microbiome and its metabolome. Cell Host Microbe 29, 394–407.e5 (2021). This human clinical trial provides promising evidence that modulation of dietary elements can aide the microbiome in recovering its metabolic capacity following broad-spectrum antibiotic treatment.
- 109.Lindell AE, Zimmermann-Kogadeeva M & Patil KR Multimodal interactions of drugs, natural compounds and pollutants with the gut microbiota. Nat. Rev. Microbiol 20, 431–443 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Vich Vila A et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun 11, 362 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R & Goodman AL Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 570, 462–467 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Javdan B et al. Personalized mapping of drug metabolism by the human gut microbiome. Cell 181, 1661–1679.e22 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Maier L et al. Unravelling the collateral damage of antibiotics on gut bacteria. Nature 599, 120–124 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.de Nies L et al. Evolution of the murine gut resistome following broad-spectrum antibiotic treatment. Nat. Commun 13, 2296 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Looft T et al. In-feed antibiotic effects on the swine intestinal microbiome. Proc. Natl Acad. Sci. USA 109, 1691–1696 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Duan Y et al. Gut resistomes, microbiota and antibiotic residues in Chinese patients undergoing antibiotic administration and healthy individuals. Sci. Total Environ 705, 135674 (2020). [DOI] [PubMed] [Google Scholar]
- 117.D’Souza AW et al. Cotrimoxazole prophylaxis increases resistance gene prevalence and alpha-diversity but decreases beta-diversity in the gut microbiome of HIV-exposed, uninfected infants. Clin. Infect. Dis doi: 10.1093/cid/ciz1186 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Thanert R et al. Antibiotic-driven intestinal dysbiosis in pediatric short bowel syndrome is associated with persistently altered microbiome functions and gut-derived bloodstream infections. Gut Microbes 13, 1940792 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kokai-Kun JF et al. Ribaxamase, an orally administered β-lactamase, diminishes changes to acquired antimicrobial resistance of the gut resistome in patients treated with ceftriaxone. Infect. Drug. Resist 13, 2521–2535 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Li J et al. Antibiotic treatment drives the diversification of the human gut resistome. Genomics Proteom. Bioinforma 17, 39–51 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lofmark S, Jernberg C, Jansson JK & Edlund C Clindamycin-induced enrichment and long-term persistence of resistant Bacteroides spp. and resistance genes. J. Antimicrob. Chemother 58, 1160–1167 (2006). [DOI] [PubMed] [Google Scholar]
- 122.Jakobsson HE et al. Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome. PLoS ONE 5, e9836 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Oldenburg CE et al. Gut resistome after oral antibiotics in preschool children in Burkina Faso: a randomized, controlled trial. Clin. Infect. Dis 70, 525–527 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Jernberg C, Lofmark S, Edlund C & Jansson JK Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J 1, 56–66 (2007). [DOI] [PubMed] [Google Scholar]
- 125.Zhang L, Huang Y, Zhou Y, Buckley T & Wang HH Antibiotic administration routes significantly influence the levels of antibiotic resistance in gut microbiota. Antimicrob. Agents Chemother 57, 3659–3666 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gasparrini AJ et al. Persistent metagenomic signatures of early-life hospitalization and antibiotic treatment in the infant gut microbiota and resistome. Nat. Microbiol 4, 2285–2297 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Johnson TA et al. Clusters of antibiotic resistance genes enriched together stay together in swine agriculture. mBio 7, e02214–e02215 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Rajer F & Sandegren L The role of antibiotic resistance genes in the fitness cost of multiresistance plasmids. mBio doi: 10.1128/mbio.03552-21 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Esaiassen E et al. Effects of probiotic supplementation on the gut microbiota and antibiotic resistome development in preterm infants. Front. Pediatr 6, 347 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Rahman SF, Olm MR, Morowitz MJ & Banfield JF Machine learning leveraging genomes from metagenomes identifies influential antibiotic resistance genes in the infant gut microbiome. mSystems doi: 10.1128/mSystems.00123-17 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Papp M & Solymosi N Review and comparison of antimicrobial resistance gene databases. Antibiotics doi: 10.3390/antibiotics11030339 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lal Gupta C, Kumar Tiwari R & Cytryn E Platforms for elucidating antibiotic resistance in single genomes and complex metagenomes. Env. Int 138, 105667 (2020). [DOI] [PubMed] [Google Scholar]
- 133.Xavier BB et al. Consolidating and exploring antibiotic resistance gene data resources. J. Clin. Microbiol 54, 851–859 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Alcock BP et al. CARD 2020: antibiotic resistome surveillance with the comprehensive antibiotic resistance database. Nucleic Acids Res 48, D517–D525 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Doster E et al. MEGARes 2.0: a database for classification of antimicrobial drug, biocide and metal resistance determinants in metagenomic sequence data. Nucleic Acids Res 48, D561–D569 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Mahmud B, Boolchandani M, Patel S & Dantas G Functional metagenomics to study antibiotic resistance. Methods Mol. Biol 2601, 379–401 (2023). [DOI] [PubMed] [Google Scholar]
- 137.Goren MG et al. Transfer of carbapenem-resistant plasmid from Klebsiella pneumoniae ST258 to Escherichia coli in patient. Emerg. Infect. Dis 16, 1014–1017 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Kent AG, Vill AC, Shi Q, Satlin MJ & Brito IL Widespread transfer of mobile antibiotic resistance genes within individual gut microbiomes revealed through bacterial Hi-C. Nat. Commun 1, 4379 (2020). This study describes the implementation of Hi-C towards elucidating HGT networks in microbial communities.
- 139.Groussin M et al. Elevated rates of horizontal gene transfer in the industrialized human microbiome. Cell 184, 2053–2067.e18 (2021). [DOI] [PubMed] [Google Scholar]
- 140.Stecher B et al. Gut inflammation can boost horizontal gene transfer between pathogenic and commensal Enterobacteriaceae. Proc. Natl Acad. Sci. USA 109, 1269–1274 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Charpentier X, Polard P & Claverys JP Induction of competence for genetic transformation by antibiotics: convergent evolution of stress responses in distant bacterial species lacking SOS. Curr. Opin. Microbiol 15, 570–576 (2012). [DOI] [PubMed] [Google Scholar]
- 142.Slager J, Kjos M, Attaiech L & Veening JW Antibiotic-induced replication stress triggers bacterial competence by increasing gene dosage near the origin. Cell 157, 395–406 (2014). [DOI] [PubMed] [Google Scholar]
- 143.Prudhomme M, Attaiech L, Sanchez G, Martin B & Claverys JP Antibiotic stress induces genetic transformability in the human pathogen Streptococcus pneumoniae. Science 313, 89–92 (2006). [DOI] [PubMed] [Google Scholar]
- 144. Modi SR, Lee HH, Spina CS & Collins JJ Antibiotic treatment expands the resistance reservoir and ecological network of the phage metagenome. Nature 499, 219–222 (2013). In this study, the authors demonstrate that antibiotic treatment results in the expansion of the frequency of phage–bacteria interactions, resulting in broader dissemination of phage-encoded antimicrobial resistance genes.
- 145.Jutkina J, Marathe NP, Flach CF & Larsson DGJ Antibiotics and common antibacterial biocides stimulate horizontal transfer of resistance at low concentrations. Sci. Total Environ 616–617, 172–178 (2018). [DOI] [PubMed] [Google Scholar]
- 146.Cairns J et al. Ecology determines how low antibiotic concentration impacts community composition and horizontal transfer of resistance genes. Commun. Biol 1, 35 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Barlow M What antimicrobial resistance has taught us about horizontal gene transfer. Methods Mol. Biol 532, 397–411 (2009). [DOI] [PubMed] [Google Scholar]
- 148.Huddleston JR Horizontal gene transfer in the human gastrointestinal tract: potential spread of antibiotic resistance genes. Infect. Drug Resist 7, 167–176 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Quince C, Walker AW, Simpson JT, Loman NJ & Segata N Shotgun metagenomics, from sampling to analysis. Nat. Biotechnol 35, 833–844 (2017). [DOI] [PubMed] [Google Scholar]
- 150.Maguire F et al. Metagenome-assembled genome binning methods with short reads disproportionately fail for plasmids and genomic Islands. Microb. Genom doi: 10.1099/mgen.0.000436 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Brown CL et al. Critical evaluation of short, long, and hybrid assembly for contextual analysis of antibiotic resistance genes in complex environmental metagenomes. Sci. Rep 11, 3753 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Moss EL, Maghini DG & Bhatt AS Complete, closed bacterial genomes from microbiomes using nanopore sequencing. Nat. Biotechnol 38, 701–707 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Lu J et al. Triclosan at environmentally relevant concentrations promotes horizontal transfer of multidrug resistance genes within and across bacterial genera. Environ. Int 121, 1217–1226 (2018). [DOI] [PubMed] [Google Scholar]
- 154.Jutkina J, Rutgersson C, Flach CF & Joakim Larsson DG An assay for determining minimal concentrations of antibiotics that drive horizontal transfer of resistance. Sci. Total Environ 548–549, 131–138 (2016). [DOI] [PubMed] [Google Scholar]
- 155.Beaber JW, Hochhut B & Waldor MK SOS response promotes horizontal dissemination of antibiotic resistance genes. Nature 427, 72–74 (2004). [DOI] [PubMed] [Google Scholar]
- 156.Zhang PY et al. Combined treatment with the antibiotics kanamycin and streptomycin promotes the conjugation of Escherichia coli. FEMS Microbiol. Lett 348, 149–156 (2013). [DOI] [PubMed] [Google Scholar]
- 157.Wang Y et al. Antiepileptic drug carbamazepine promotes horizontal transfer of plasmid-borne multi-antibiotic resistance genes within and across bacterial genera. ISME J 13, 509–522 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Handel N, Otte S, Jonker M, Brul S & ter Kuile BH Factors that affect transfer of the IncI1 β-lactam resistance plasmid pESBL-283 between E. coli strains. PLoS ONE 10, e0123039 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Johnsen AR & Kroer N Effects of stress and other environmental factors on horizontal plasmid transfer assessed by direct quantification of discrete transfer events. FEMS Microbiol. Ecol 59, 718–728 (2007). [DOI] [PubMed] [Google Scholar]
- 160.Lopatkin AJ et al. Antibiotics as a selective driver for conjugation dynamics. Nat. Microbiol 1, 16044 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Feld L et al. Selective pressure affects transfer and establishment of a Lactobacillus plantarum resistance plasmid in the gastrointestinal environment. J. Antimicrob. Chemother 61, 845–852 (2008). [DOI] [PubMed] [Google Scholar]
- 162.Lopatkin AJ, Sysoeva TA & You L Dissecting the effects of antibiotics on horizontal gene transfer: analysis suggests a critical role of selection dynamics. Bioessays 38, 1283–1292 (2016). In this examination of the reports on the effects of antibiotics on conjugation, the authors make a compelling case for how antibiotic selection confounds the experimental results and their interpretation. The authors call for more careful experimental design that enables decoupling of the antibiotic effects on conjugation and bacterial growth rates. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Deng F, Li Y & Zhao J The gut microbiome of healthy long-living people. Aging 11, 289–290 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Connelly S et al. SYN-004 (ribaxamase), an oral beta-lactamase, mitigates antibiotic-mediated dysbiosis in a porcine gut microbiome model. J. Appl. Microbiol 123, 66–79 (2017). [DOI] [PubMed] [Google Scholar]
- 165.de Gunzburg J et al. Protection of the human gut microbiome from antibiotics. J. Infect. Dis 217, 628–636 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wan MLY, Forsythe SJ & El-Nezami H Probiotics interaction with foodborne pathogens: a potential alternative to antibiotics and future challenges. Crit. Rev. Food Sci. Nutr 59, 3320–3333 (2019). [DOI] [PubMed] [Google Scholar]
- 167.Drolia R et al. Receptor-targeted engineered probiotics mitigate lethal Listeria infection. Nat. Commun 11, 6344 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Koh E et al. Engineering probiotics to inhibit Clostridioides difficile infection by dynamic regulation of intestinal metabolism. Nat. Commun 13, 3834 (2022). This paper reports on the engineering of E. coli Nissle 1917 to selectively secrete a bile salt hydrolase in a dysbiotic environment, restoring the intestinal bile salt metabolism and impeding C. difficile germination and disease.
- 169.Ajami NJ, Cope JL, Wong MC, Petrosino JF & Chesnel L Impact of oral fidaxomicin administration on the intestinal microbiota and susceptibility to Clostridium difficile colonization in mice. Antimicrob. Agents Chemother doi: 10.1128/AAC.02112-17 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Brochado AR et al. Species-specific activity of antibacterial drug combinations. Nature 559, 259–263 (2018). In this paper, the authors demonstrate that the combined activity of an antibiotic combination is commonly species specific and strain specific, providing evidence for the possibility of developing narrow-spectrum therapies based on drug combinations.
- 171.Stentz R et al. Cephalosporinases associated with outer membrane vesicles released by Bacteroides spp. protect gut pathogens and commensals against β-lactam antibiotics. J. Antimicrob. Chemother 70, 701–709 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Kokai-Kun JF et al. Use of ribaxamase (SYN-004), a β-lactamase, to prevent Clostridium difficile infection in β-lactam-treated patients: a double-blind, phase 2b, randomised placebo-controlled trial. Lancet Infect. Dis 19, 487–496 (2019). [DOI] [PubMed] [Google Scholar]
- 173. Cubillos-Ruiz A et al. An engineered live biotherapeutic for the prevention of antibiotic-induced dysbiosis. Nat. Biomed. Eng doi: 10.1038/s41551-022-00871-9 (2022). The authors of this paper generate an engineered L. lactis strain that degrades intestinal β-lactams through secretion of a heterodimeric β-lactamase. Notably, given its split nature and extracellular assembly, the β-lactamase protects the microbial community from β-lactams but does not confer resistance to the host L. lactis.
- 174.Vehreschild M et al. An open randomized multicentre phase 2 trial to assess the safety of DAV132 and its efficacy to protect gut microbiota diversity in hospitalized patients treated with fluoroquinolones. J. Antimicrob. Chemother 77, 1155–1165 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Burdet C et al. Protection of hamsters from mortality by reducing fecal moxifloxacin concentration with dav131a in a model of moxifloxacin-induced Clostridium difficile colitis. Antimicrob. Agents Chemother doi: 10.1128/AAC.00543-17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.de Gunzburg J et al. Targeted adsorption of molecules in the colon with the novel adsorbent-based medicinal product, DAV132: a proof of concept study in healthy subjects. J. Clin. Pharmacol 55, 10–16 (2015). [DOI] [PubMed] [Google Scholar]
- 177.Hryckowian AJ et al. Microbiota-accessible carbohydrates suppress Clostridium difficile infection in a murine model. Nat. Microbiol 3, 662–669 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.McDonald JAK et al. Inhibiting growth of Clostridioides difficile by restoring valerate, produced by the intestinal microbiota. Gastroenterology 155, 1495–1507.e15 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Pruss KM et al. Mucin-derived O-glycans supplemented to diet mitigate diverse microbiota perturbations. ISME J 15, 577–591 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Schnizlein MK, Vendrov KC, Edwards SJ, Martens EC & Young VB Dietary xanthan gum alters antibiotic efficacy against the murine gut microbiota and attenuates Clostridioides difficile colonization. mSphere doi: 10.1128/mSphere.00708-19 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Simpson HL et al. Soluble non-starch polysaccharides from plantain (Musa × paradisiaca L.) diminish epithelial impact of Clostridioides difficile. Front. Pharmacol 12, 766293 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Roberts CL et al. Soluble plantain fibre blocks adhesion and M-cell translocation of intestinal pathogens. J. Nutr. Biochem 24, 97–103 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Button JE et al. Dosing a synbiotic of human milk oligosaccharides and B. infantis leads to reversible engraftment in healthy adult microbiomes without antibiotics. Cell Host Microbe 30, 712–725.e7 (2022). [DOI] [PubMed] [Google Scholar]
- 184.Ghani R, Mullish BH, Roberts LA, Davies FJ & Marchesi JR The potential utility of fecal (or intestinal) microbiota transplantation in controlling infectious diseases. Gut Microbes 14, 2038856 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Suez J et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174, 1406–1423.e16 (2018). [DOI] [PubMed] [Google Scholar]
- 186.Orenstein R et al. Durable reduction of Clostridioides difficile infection recurrence and microbiome restoration after treatment with RBX2660: results from an open-label phase 2 clinical trial. BMC Infect. Dis 22, 245 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kao D et al. The effect of a microbial ecosystem therapeutic (MET-2) on recurrent Clostridioides difficile infection: a phase 1, open-label, single-group trial. Lancet Gastroenterol. Hepatol 6, 282–291 (2021). [DOI] [PubMed] [Google Scholar]
- 188.Feuerstadt P et al. SER-109, an oral microbiome therapy for recurrent Clostridioides difficile infection. N. Engl. J. Med 386, 220–229 (2022). [DOI] [PubMed] [Google Scholar]
- 189. Dsouza M et al. Colonization of the live biotherapeutic product VE303 and modulation of the microbiota and metabolites in healthy volunteers. Cell Host Microbe 30, 583–598.e8 (2022). This study is the first clinical demonstration of the therapeutic efficacy and safety of a rationally defined bacterial consortium developed to treat recurrent C. difficile infections.
- 190.Francis MB, Allen CA, Shrestha R & Sorg JA Bile acid recognition by the Clostridium difficile germinant receptor, CspC, is important for establishing infection. PLoS Pathog 9, e1003356 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Winston JA & Theriot CM Impact of microbial derived secondary bile acids on colonization resistance against Clostridium difficile in the gastrointestinal tract. Anaerobe 41, 44–50 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Kwak S et al. Impact of investigational microbiota therapeutic RBX2660 on the gut microbiome and resistome revealed by a placebo-controlled clinical trial. Microbiome 8, 125 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.DeFilipp Z et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N. Engl. J. Med 381, 2043–2050 (2019). [DOI] [PubMed] [Google Scholar]
- 194.Zellmer C et al. Shiga toxin-producing Escherichia coli transmission via fecal microbiota transplant. Clin. Infect. Dis 72, e876–e880 (2021). [DOI] [PubMed] [Google Scholar]
- 195.Guarner F & Schaafsma GJ Probiotics. Int. J. Food Microbiol 39, 237–238 (1998). [DOI] [PubMed] [Google Scholar]
- 196.Montassier E et al. Probiotics impact the antibiotic resistance gene reservoir along the human GI tract in a person-specific and antibiotic-dependent manner. Nat. Microbiol 6, 1043–1054 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Zuo F, Chen S & Marcotte H Engineer probiotic bifidobacteria for food and biomedical applications — current status and future prospective. Biotechnol. Adv 45, 107654 (2020). [DOI] [PubMed] [Google Scholar]
- 198.Goh YJ & Barrangou R Harnessing CRISPR–Cas systems for precision engineering of designer probiotic lactobacilli. Curr. Opin. Biotechnol 56, 163–171 (2019). [DOI] [PubMed] [Google Scholar]
- 199.Kwak S, Mahmud B & Dantas G A tunable and expandable transactivation system in probiotic yeast Saccharomyces boulardii. ACS Synth. Biol 11, 508–514 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Gelfat I et al. Single domain antibodies against enteric pathogen virulence factors are active as curli fiber fusions on probiotic E. coli Nissle 1917. PLoS Pathog 18, e1010713 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Forkus B, Ritter S, Vlysidis M, Geldart K & Kaznessis YN Antimicrobial probiotics reduce Salmonella enterica in turkey gastrointestinal tracts. Sci. Rep 7, 40695 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Tscherner M, Giessen TW, Markey L, Kumamoto CA & Silver PA A synthetic system that senses Candida albicans and inhibits virulence factors. ACS Synth. Biol 8, 434–444 (2019). [DOI] [PubMed] [Google Scholar]
- 203.Jayaraman P, Holowko MB, Yeoh JW, Lim S & Poh CL Repurposing a two-component system-based biosensor for the killing of Vibrio cholerae. ACS Synth. Biol 6, 1403–1415 (2017). [DOI] [PubMed] [Google Scholar]
- 204.Hwang IY et al. Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat. Commun 8, 15028 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Palmer JD et al. Engineered probiotic for the inhibition of salmonella via tetrathionate-induced production of microcin H47. ACS Infect. Dis 4, 39–45 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Willmann M et al. Antibiotic selection pressure determination through sequence-based metagenomics. Antimicrob. Agents Chemother 59, 7335–7345 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Buelow E et al. Effects of selective digestive decontamination (SDD) on the gut resistome. J. Antimicrob. Chemother 69, 2215–2223 (2014). [DOI] [PubMed] [Google Scholar]
- 208.Mirzayi C et al. Reporting guidelines for human microbiome research: the STORMS checklist. Nat. Med 27, 1885–1892 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Brewster R et al. Surveying gut microbiome research in Africans: toward improved diversity and representation. Trends Microbiol 27, 824–835 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Stokholm J et al. Antibiotic use during pregnancy alters the commensal vaginal microbiota. Clin. Microbiol. Infect 20, 629–635 (2014). [DOI] [PubMed] [Google Scholar]
- 211.Dubos R, Schaedler RW & Stephens M The effect of antibacterial drugs on the fecal flora of mice. J. Exp. Med 117, 231–243 (1963). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Hertz FB et al. Effects of antibiotics on the intestinal microbiota of mice. Antibiotics doi: 10.3390/antibiotics9040191 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Zhang Y, Limaye PB, Renaud HJ & Klaassen CD Effect of various antibiotics on modulation of intestinal microbiota and bile acid profile in mice. Toxicol. Appl. Pharmacol 277, 138–145 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Sun L et al. Antibiotic-induced disruption of gut microbiota alters local metabolomes and immune responses. Front. Cell Infect. Microbiol 9, 99 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Burdet C et al. Ceftriaxone and cefotaxime have similar effects on the intestinal microbiota in human volunteers treated by standard-dose regimens. Antimicrob. Agents Chemother doi: 10.1128/AAC.02244-18 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Tirelle P et al. Comparison of different modes of antibiotic delivery on gut microbiota depletion efficiency and body composition in mouse. BMC Microbiol 20, 340 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Stokholm J, Sevelsted A, Bonnelykke K & Bisgaard H Maternal propensity for infections and risk of childhood asthma: a registry-based cohort study. Lancet Respir. Med 2, 631–637 (2014). [DOI] [PubMed] [Google Scholar]
- 218.Tao C, Zhang Q, Zeng W, Liu G & Shao H The effect of antibiotic cocktails on host immune status is dynamic and does not always correspond to changes in gut microbiota. Appl. Microbiol. Biotechnol 104, 4995–5009 (2020). [DOI] [PubMed] [Google Scholar]
- 219.Lavelle A et al. Baseline microbiota composition modulates antibiotic-mediated effects on the gut microbiota and host. Microbiome 7, 111 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Akbar S et al. Changes in the life history traits of Daphnia magna are associated with the gut microbiota composition shaped by diet and antibiotics. Sci. Total Environ 705, 135827 (2020). [DOI] [PubMed] [Google Scholar]
- 221.Harrison CA et al. Sexual dimorphism in the response to broad-spectrum antibiotics during T cell-mediated colitis. J. Crohns Colitis 13, 115–126 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Ruczizka U et al. Early parenteral administration of ceftiofur has gender-specific short- and long-term effects on the fecal microbiota and growth in pigs from the suckling to growing phase. Animals doi: 10.3390/ani10010017 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Fujisaka S et al. Antibiotic effects on gut microbiota and metabolism are host dependent. J. Clin. Invest 126, 4430–4443 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Jansen K, Pou Casellas C, Groenink L, Wever KE & Masereeuw R Humans are animals, but are animals human enough? A systematic review and meta-analysis on interspecies differences in renal drug clearance. Drug Discov. Today 25, 706–717 (2020). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
