SUMMARY
Communities of microorganisms (microbiota) are present in all habitats on Earth and are relevant for agriculture, health, and climate. Deciphering the mechanisms that determine microbiota dynamics and functioning within the context of their respective environments or hosts (the microbiomes) is crucially important. However, the sheer taxonomic, metabolic, functional, and spatial complexity of most microbiomes poses substantial challenges to advancing our knowledge of these mechanisms. While nucleic acid sequencing technologies can chart microbiota composition with high precision, we mostly lack information about the functional roles and interactions of each strain present in a given microbiome. This limits our ability to predict microbiome function in natural habitats and, in the case of dysfunction or dysbiosis, to redirect microbiomes onto stable paths. Here, we will discuss a systematic approach (dubbed the N+1/N−1 concept) to enable step-by-step dissection of microbiome assembly and functioning, as well as intervention procedures to introduce or eliminate one particular microbial strain at a time. The N+1/N−1 concept is informed by natural invasion events and selects culturable, genetically accessible microbes with well-annotated genomes to chart their proliferation or decline within defined synthetic and/or complex natural microbiota. This approach enables harnessing classical microbiological and diversity approaches, as well as omics tools and mathematical modeling to decipher the mechanisms underlying N+1/N−1 microbiota outcomes. Application of this concept further provides stepping stones and benchmarks for microbiome structure and function analyses and more complex microbiome intervention strategies.
KEYWORDS: microbiota, microbiome development, focal strains, inoculants, modeling, systems’ analysis
INTRODUCTION: WHY MICROBIOME ENGINEERING?
Microbial communities, or microbiota, self-organize in a seemingly spontaneous way within the spatial, temporal, physical, chemical, and biological boundary conditions of their environments or hosts (i.e., their habitats). The microbiota within the context of these boundaries (i.e., “microbiomes,” Fig. 1A, Box 1) (1) contribute to important ecological and biogeochemical processes (2), as well as to plant (3), human (4), and animal health (5–8). Alarmingly, an increasing body of knowledge has pointed to recent changes in microbiome functioning [for example, loss of diversity or functions (9, 10)] across all systems (e.g., soils, animals, and human guts) (11–13). From a human perspective, this may have potentially dire consequences for public health, agricultural production, and environmental quality. Underlying causes may include a variety of external factors and conditions, such as pollution, climate change, land and agricultural management, travel to foreign areas, extensive use of antimicrobial compounds, pharmaceuticals, and nutritional habits. These changes motivate a deeper understanding of microbiome structure and function across systems in order to potentially restore and maintain ecosystem function.
Fig 1.
Concept terms in N+1/N−1 microbiome interventions. (A) All habitats (environments and hosts) are occupied by microbiota, (different) assemblages of microbial cells and taxa, collectively forming the microbiomes, as illustrated. WWTP, Wastewater treatment plant. (B) N+1/N−1 approaches intend to attain a state into which is introduced (+1) or eliminated (−1) a microbial taxon (focal strains) from the resident microbiota with its N (total) taxa, to study their outcomes, and understand the molecular and ecological mechanisms leading to the observed microbiome’s properties. (C) Examples of specific applied N+1/N−1 intervention goals, to (D) e.g., complement microbiome functions in case of pollution damage, or restore functional networks by reducing pathogen loads. Part D modified after ref. (14).
BOX1: Glossary of terms.
Microbiota / microbial community: the collective (living) microorganisms in a habitat (including Bacteria, Archaea, and eukaryotic microbes). Generally not thought to include viruses, phages, or naked DNA (1).
Microbiome: the ensemble of all microbes integrated within the context of its habitat boundaries (environmental system or host); microbiota and their “theatre of activity: structural elements, metabolites/signal molecules, and the surrounding environmental conditions” (1).
Habitat: the array of biotic and abiotic factors, spatial constraints, physico-chemical conditions, and dynamic properties (flow, fluctuations) that characterize an environment or host.
Dispersal: flow of microorganisms into and out of a particular habitat (15, 16).
(Evolutionary) drift: random shifts in the relative abundance of taxa or genotypes.
Diversification: genetic change by horizontal gene transfer or mutation.
Niche: a set of biotic and abiotic conditions that determine the proliferation of a microbial cell or taxa in a given habitat. To differentiate: the potential and the realized niche (17).
Probiotic: live microorganism that, when administered in adequate amounts, may confer a health benefit on the host.
Prebiotic: substrate which is selective for the proliferation or maintenance of the probiotic within the target microbiome, either directly benefiting the probiotic or indirectly producing favorable niche conditions.
Synbiotic: combination of pro- and prebiotic(s).
Postbiotic: preparation of inanimate microorganisms and/or their components with a claimed health benefit on the host. Effective postbiotics must contain inactivated microbial cells or cell components with or without metabolites that contribute to observed health benefits.
Focal strain: a single microbial strain to be studied for its proliferation, activity within, or effects on a microbiota.
N+1 engineering: intervention in a microbiome of choice to add a focal strain, either for permanent engraftment, for temporary functional effects or for influencing the developmental trajectory of the microbiome.
N−1 engineering: intervention in a microbiome of choice to remove a focal strain, either to diminish its immediate effects (e.g., an infection) or to prevent its influence on the developmental trajectory of the microbiome.
Advances in sequencing and bioinformatic tools have enabled unprecedented insights into the taxonomic and functional richness of microbiota (18, 19). However, their collective properties remain largely unresolved, leading to several important unanswered questions: how do external factors disrupt the tendency of natural communities to maintain compositional and functional homeostasis (10), and alter the state, the maintenance, the resilience, or temporal dynamics (“trajectories”) of microbiomes? How can negative consequences on microbiome homeostasis be recognized, predicted, prevented, and, possibly, reverted? To rationally intervene in disrupted or dysfunctional microbiomes, we need a much better understanding of the processes that lead to their formation and maintenance, as well as experience using empirical tools that we might eventually deploy to invoke an intended recovery or stabilization. Currently, complex microbiome engineering (20) that would involve simultaneously modulating many taxa or factors is likely unfeasible. These challenges, therefore, pose the question of whether a step-by-step approach of learning from single focal strains—one that is rooted in established expertise with cultivable microbes in agricultural (21), environmental (22), nutritional (23, 24), and pathogen research (25) can provide the insights necessary to enable microbiome engineering.
Here, we discuss an iterative approach to rationally intervene in microbiomes and decipher general principles underlying microbiome assembly and functioning. We call this approach the N+1/N−1 concept, a framework to add or suppress individual microbiota members (focal strains) within the context of their habitat, and to probe outcomes and underlying functional processes (Fig. 1B). In this sense, N+1/N−1 can also be considered as an intended state to be achieved, as well as a starting point for developing more complex microbiome engineering strategies (Fig. 1C). Many of our ideas center around deploying single strains to, for example, enable specific system functionalities; or eliminate single strains (e.g., recurring pathogens; Fig. 1D). Through successive cycles of designing, testing, and measuring (e.g., by removing or adding different strains, or altering nutrient profiles or habitat conditions), we can learn from ostensibly simple N+1/N−1 intervention tasks to improve our capability to rationally intervene in dysfunctional microbiomes.
This review begins with a detailed explanation of the N+1/N−1 concept; followed by discussions of how N+1/N−1 studies can be envisioned and designed, which tools are available to measure and quantify microbiome processes, and how computational models can be leveraged to understand the effects at the microbiome’s system level. Since N+1/N−1 approaches are intimately related to natural processes of strain dispersal, we will then summarize what we might learn from natural N+1/N−1 occurrences to improve methods for microbiome interventions. Finally, we will present a number of case studies on N+1/N−1 intervention examples to summarize what has been learned, and how to go forward. Throughout, we emphasize the importance of systematic approaches across a broad spectrum of host- and environmental microbiomes to delineate general principles of microbiome intervention and differentiate them from system-specific traits.
THE N+1/N−1 CONCEPT
Precise design across a broad spectrum of host and environmental microbiomes
The N+1/N−1 concept is simple in its basic definition: to introduce (“+1”) or eliminate (“−1”) a focal strain of interest within a background targeted microbiome composed of N taxa (Fig. 1B). There may be various reasons (from an intervention point of view) to achieve N+1/N−1 states. For example, so-called suppressive soils have a lower incidence of agricultural plant diseases, which has been attributed to higher abundances of plant-beneficial bacteria (26). One might thus aim to complement disease-permissive soils with a specific plant-beneficial bacterial strain in order to ward off plant pathogens and enhance plant growth without intervention with phytochemicals (27) (Fig. 1C). As another example, one might strive to specifically remove a pathogen from the human gut after infection without the use of antibiotics (28), or to restore contaminated soils with detoxifying bacterial strains (29, 30).
Though simple, this N+1/N−1 concept opens the door to investigating the numerous experimental considerations and complex ecological processes that arise in microbiome engineering. For example, does introducing a focal strain into a microbiota mean that the strain should become permanently engrafted in that microbiota? Depending on the intervention goal, it might be sufficient to achieve only transient establishment(s), during which the focal strain can deploy its functionalities and then disappear. Depending on the dynamics or characteristics of the microbiome under scrutiny, adding or removing a focal strain may imply multiple successive interventions, in order to interfere with the microbiome development or its homeostatic processes. More generally, when aiming to introduce a strain that is beneficial to a host, it is critical to first understand how it disperses and proliferates in its habitat. This understanding in turn requires knowledge on the strain’s niche, its competitors and predators, and when and where it best deploys its beneficial functionalities.
Indeed, strain dispersal, survival, proliferation, and decline are often multifactorial processes, which depend on the strain’s genetic makeup, the microbiota composition, interspecific interactions, and characteristics of the habitat and niche. To generalize our understanding of these processes, N+1/N−1 studies present a tractable way to profile a wide range of different microbiomes and habitats (including those without and with hosts). As such, they can span both descriptive and mechanistic studies with native microbiomes as well as synthetic communities of reduced complexity that reproduce native microbiomes. Synthetic communities of reduced complexity (compared to natural microbiomes) have the advantage of better reproducibility, allowing more precise bottom-up in vitro systems control over the N+1/N−1 (knock-in and knock-out) composition and potential outcomes [see, for example, recent work on plant leaf synthetic microbiota (14)]. Ideally, such synthetic communities reproduce a native microbiome counterpart, and their composition can be guided by inference from multi-omics analysis of native microbiomes to have appropriate taxonomic and functional representation.
Importantly, N+1/N−1 approaches can bridge the gap between correlative and descriptive studies of microbiome composition (which are typically limited in their taxonomic and mechanistic resolution) and molecular studies focusing on causality in simplified systems (which improve reproducibility but may be biased by the composition of its microbial members). When considering the focal strains selected, an N+1/N−1 approach can more readily allow one to establish the molecular basis of their growth or demise within their respective microbiomes. The resulting mechanistic and ecological knowledge can then be extended by predicting the outcome of microbiome interventions. Failure to predict such outcomes would help to identify crucial remaining knowledge gaps and encourage the re-examining of established hypotheses. Conversely, successful predictions would drive the improvement of precision microbiome intervention tools.
N+1/N−1 experimental design
The first step in an N+1/N−1 study is the selection of focal strains. Ideal candidates are strains of relevance for a future application or therapy (e.g., plant-growth-promoting bacteria, probiotics, or xenobiotic metabolic complements; Fig. 1C) but could also consist of a presumed keystone taxon in a microbiota interaction network (31–33) or sentinel species (34). Interventions focused on pathogenic strains have the goal not to introduce the focal strain, but rather to remove it from a microbiome (N−1). Nonetheless, infective pathogens can also be studied through an N+1 framing, as they have evolved specifically to invade microbiomes. Understanding this invasive behavior may help to design strategies that lead to the pathogen’s suppression or removal. For example, non-pathogenic strains that can fill the same niche may inhibit the pathogen from proliferating within the microbiome (35–38). We may also learn from pathogen invasion to guide strategies to improve the transient establishment of a (non-pathogenic) N+1 inoculant. Additional considerations in the selection of a focal strain may include genetic tractability (to enable comparison with microbiota properties in the presence of a mutated focal strain) and a good experience basis for physiological and functional screening (Fig. 2). Additionally, focal strains should minimize potential biological safety concerns, especially in the context of health or environmental intervention studies.
Fig 2.
The who-and-how of N+1/N−1 microbiome interventions. Focal strains (symbolized by the blue circle) to be added into or removed from the target habitat (dark brackets) and its microbiota (N, represented here as a taxa co-occurrence network of positive: blue, and negative: red, connecting lines). Experimental designs need at least three conditions; the focal strain alone in the habitat (+1), the resident microbiota alone (N), and the inoculation condition (N+1). Examples of the details to define under “who,” “how,” etc. are explained in the main text. Iteration of the approach results from, e.g., follow-up experiments with mutant focal strains or altered habitat conditions and microbiota, in order to uncover molecular, ecological, or mechanistic processes.
Any N+1/N−1 study needs a minimum of four components: the focal strain by itself, the resident microbiota by itself, the combination of the focal strain within the resident microbiota, and a defined habitat (Fig. 2). By comparing the growth of the focal strain in isolation with its growth in the presence of the resident microbiota, kinetic and physiological differences can be assessed, which are the basis for uncovering underlying regulatory mechanisms, metabolic changes, and ecological processes. Under the condition that one can achieve similar growth rates and phases of the focal strain in laboratory and native microbiome conditions, such comparisons can point to specific adaptive mechanisms of the focal strain and to the prevailing host/habitat factors to which it reacts. These results can then be confirmed with focal strains mutated in identified characteristics or pathways.
The choice of resident microbiota into which the focal strain is introduced can range in complexity, different levels of which can be conducive to uncovering specific mechanisms and processes (e.g., interspecific interactions), and system-level properties (e.g., colonization resistance or niche availability) that control the focal strain’s proliferation, survival, and impact (Fig. 3A). The simplest set-ups can consist of paired co-cultures between the focal strain and (native) culturable microbiota members (39), or even with any arbitrary culturable strains relevant to specific ecological processes (e.g., competition) (40). Combined information from co-cultures can to some extent predict higher-order interactions and the dominating community network (41, 42). As a next step in complexity, one could design simplified synthetic communities with members obtained either from a collection of culturable isolates (43) or from the native microbiome (38, 44–47). The advantage of choosing simplified communities is a more controllable, reproducible, and defined microbiome, which can help to produce N−1 states by composition and dissect effects of N+1 inoculations, treatments, or other system perturbations. However, such synthetic communities remain a simplification, of which it is important to understand the differences to the native microbiota. Multiple simplified microbiota have been compiled in recent years, representing a range of habitats. These include, for example, the Oligo-MM-12 (38), Oligo-MM-19 (48), and LCM communities (49) for the mouse gut microbiome, and a defined 104-strain hCom1 human gut microbiota mixture (50). Other model animals, such as the honeybee (46, 47), have also been used to study specific gut microbiota effects. Simplified representative synthetic communities have also been produced from cultured isolates for complex microbiomes that are inherently open in nature, like plants or topsoil. Examples here include the At-SPHERE [5 phyla, 430 plant leaf and root isolates; reference (44)], or a 21-member (4 phyla) collection of soil isolates (45). Higher complexity can be achieved by cell “washes” from the natural habitat of the microbiota and their controlled reintroduction, but this comes at the loss of some reproducibility (45).
Fig 3.
Potential lay-outs of N+1/N−1 studies. (A) Focal strains (here in blue) can be tested in conjunction with microbiota of increasing complexity to establish ecological and molecular mechanisms of their potential survival and proliferation, as well as their impact on the microbiome in the short or longer term. (B) Culturing systems can be tuned specifically to the main study objectives and/or methodological tools available.
The final system choice to make with the selected focal strain(s) and resident communities is the habitat itself (Fig. 3B). In addition to the native habitat of the resident microbiota, one can test a range of different culturing conditions that allow for more precise control of the habitat and comparison across systems to identify consistent underlying mechanisms and processes. For example, liquid-suspended culturing is accessible and offers flexibility in terms of nutrients and growth conditions. It is also simple to parallelize in medium- to high throughput while maintaining flexibility to analyze microbiota (compositional) outcomes. At higher throughput, one could choose for fragmented and miniaturized growth in, e.g., encapsulated beads (51), microdroplets (41), or microchip formats (42). Spatial constraints are more easily implemented in microfluidic chambers (52–54), on surfaces (55), or “reprints” of native habitat structures (56).
In case the native habitat is preferred, one could reproduce this in a sterile form with the same overall conditions. When working with animal or plant hosts, one could deploy gnotobiotic or microbe-depleted individuals. This allows a controlled recolonization of the habitats, which is an extremely important element of N+1/N−1 studies. For example, gnotobiotic animal models permit growing microbiomes with known strain composition; in which case the natural habitat (e.g., animal gut) is colonized by a resident microbiota with reduced complexity. Cross-inoculation experiments are frequently possible, as demonstrated by human fecal microbiota grown in gnotobiotic mice as surrogate hosts, so-called “humanized microbiota” mice (57). Also, microbiota-depleted bees have been successfully recolonized by mixtures of their resident gut microbiota of defined composition and reduced complexity, to test specific host-microbiota and even behavioral effects (46, 47, 58). Similarly, gnotobiotic plants permit culturing of reduced complexity leaf or root microbiomes, which has been exploited to measure the effect of individual members of the At-LSPHERE microbiota collection by drop-out experiments (59). As an alternative approach to representative synthetic communities with cultured isolates one can also extract the mixture of resident microbiota from their original habitat and transplant this mixture in reproduced sterile or gnotobiotic habitats, as recently shown for soils (45). In certain cases, this results in reproducible high-complexity communities that can serve as resident background for testing N+1/N−1 scenarios.
Methods to monitor focal strains
The proliferation and physiology of an introduced focal strain within the microbiota and selected habitat can be measured with a variety of methods. For example, high-throughput sequencing and mass-spectrometry approaches can quantify changes in gene and protein expression as well as in metabolism between defined laboratory growth conditions and natural environments or the habitats intended for targeted application (60, 61). Instead of single focal strain genotypes, random insertion (36, 60) or CRISPRi mutant libraries (62) can be produced and inoculated, in order to gain functional evidence for the relevant mechanisms enabling the focal strain to proliferate. Kept in a library format of individual clones with each a single insertion mutant or CRISPRi-inhibited transcript, the complete library covers multiple times all coding content, such that positive and negative fitness effects and their modulation by habitat conditions and the microbiome can be detected. This is typically accomplished by growing the library under the conditions of the intended application, in comparison with defined laboratory conditions. Samples taken at different incubation time points are used for DNA extraction and sequencing of the marker insertion positions or CRISPRi-targets. Subsequently, the relative abundances of all marker insertion/target positions in the different conditions are quantified and compared among each other and to those in the starting library. Depletion of marker insertion positions is interpreted as a sign of their negative fitness effect, whereas enrichment may point to fitness gain.
Fluorescently tagged focal strains may also be used to address specific questions on their in situ behavior or individual cell variability within a microbiome (63–65). Growth dilution markers [e.g., conditionally replicative plasmids (66), inducible fluorescent protein expression (67), or isotope labeling (68)], oscillation circuit-based molecular clockworks (69), and isogenic genomic barcoding tags can further inform on in situ growth rates and population bottlenecks (36, 70, 71). Fluorescent reporter systems can also reveal the actual spatial, temporal, and population-wide distribution of specific metabolic or physiological activities of the focal strain (54, 72–74), which can then be related to habitat substrate or niche availability and metabolic adaptation. Inducible off-on tags can be used to develop sentinel strains whose exposure to a specific stimulus in their habitat can be recorded and read out following their re-isolation (34, 75).
Microbiomes that can be observed over longer (e.g., evolutionary) timescales can be deployed to re-isolate clones of the focal strain during its colonization, survival, and/or population decline (76–78). The genomes of such isolates can be sequenced to identify the emergence of genetic or phenotypic traits that facilitated habitat adaptation (79). Ideally, this can be combined with in vitro growth experiments to validate assumed adaptive traits in niche colonization.
Methods to follow resident microbiota functioning and focal strain effects
Changes in the taxonomic composition of the microbiota can be quantified using amplicon sequencing of phylogenetic marker genes such as the 16S rRNA gene for Bacteria and Archaea, the 18S rRNA gene for microeukaryotes such as protists, or the internal transcribed spacer region for fungi. Depending on the compositional complexity of the resident microbiota, sequencing approaches may be replaced by, for instance, quantitative PCR. Although amplicon sequencing has the disadvantage of a lower taxonomic resolution, its convenience lies in its speed, cost efficiency per sample, and greater compositional depth. Interpretation of resident microbiota composition patterns is facilitated by numerous recent tools. By using a closed reference for mapping data (80), comparisons can be made across a large variety of studies. This can be explored in web interfaces such as the Microbe Atlas Database (https://microbeatlas.org/). The Microbe Atlas Database follows a single standardized approach to deduce taxonomic memberships, making it extremely powerful to compare taxa occurrences across habitats. Still, the current taxonomic resolution of Microbe Atlas and similar databases is limited because of their reliance on a single marker gene. In addition, only limited metabolic and functional microbiota information can be inferred from amplicon sequencing (81). Methods have been proposed to overcome this [e.g., reference (82)], but these depend on the availability of annotated reference genomes and ignore possible strain-level variations.
Alternative efforts aim to integrate taxonomic and functional information from metagenomic sequencing in the form of metagenome-assembled genomes [MAGs (83)]. As an example, mOTUs (metagenomic operational taxonomic units) is a community-benchmarked (84) metagenomic profiling tool that, in its latest version (85), leverages large-scale reconstruction of MAGs [e.g., reference (18)] and single-copy marker genes that can be identified in any metagenome (86). This makes mOTUs genome-independent, which is particularly useful for N+1/N−1 approaches involving resident microbiota that are not well represented by available reference genomes: a common issue across a wide range of environments and hosts (85). Ongoing efforts aim at mapping sequences of 16S rRNA genes and single-copy marker genes to their MAGs of origin, which would allow for quantifying gene functional compositions rather than inferring functions from taxonomic profiles, all while accounting for taxa without available reference genomes. These efforts will be facilitated by deploying long-read sequencing for metagenomics (87).
For simplified resident communities, amplicon sequencing can serve to follow compositional changes at high depth and resolution. It should be noted, however, that currently available sequencing strategies do not provide absolute but only relative strain abundances, except if combined with DNA spiking (88). Since the absolute population size is a crucial readout (e.g., for estimating growth), it can also be quantified using plate counts, flow cytometry, or quantitative PCR (89). Recent advances in flow cytometry have also demonstrated that community size quantification and population “typing” can be achieved simultaneously by applying machine learning classifiers, at least for low-complexity communities (90, 91). Such advances may greatly simplify the analysis of high-resolution time dynamic compositional changes emerging from N+1/N−1 experiments.
The analysis of diversity and compositional changes in the resident microbiota can be further coupled to pairwise random growth experiments with the inoculants, in order to understand how the focal strain may benefit or inhibit specific microbiota members. For small, defined microbial consortia, full interaction networks can be obtained by exhaustive (paired) co-culturing. Such networks are valuable tools to understand how a system is structured, contributing to hypotheses on, for example, the community-wide effects of centrally positioned microbes with high disruptive potential (31, 32, 59, 92, 93). While recent co-cultivation methods (42, 94) have enabled the screening of increasingly complex communities, obtaining networks for large, natural communities is still out of reach. Statistical methods based on co-occurrence (95–98) or time series data (99, 100) are useful instruments to start investigating such systems, allowing a first glimpse at the structure of the underlying ecological networks. More practically, they could guide the design of tractable synthetic microbial consortia—ensuring the predicted ecological network is well-covered—or help to strategically prioritize focal strains with a high predicted impact on the overall community. Schäfer et al., for example, systematically studied the impact of more than 200 individual strains on a defined synthetic community of 15 members to identify the major interacting species (39).
At a systems level, responses of the resident microbiome to the presence or absence of focal strains or to changes in biotic or abiotic factors can be followed by metatranscriptomics, metaproteomics, or meta-metabolomics. These methods are particularly applicable for observing changes in carbon or nutrient flows relevant to an N+1/N−1 engineering process (101).
Impact of spatial organization on the N+1/N−1 concept
Microbial habitats generally do not consist of homogenously mixed environments with ideally suspended individual cells of different strains but rather form highly spatially structured and dynamic assemblages of strains, biofilms, microcolonies, and with accompanying (dynamic) gradients in nutrients or electron acceptors. Unfortunately, studying spatial structures of complex microbiota is a challenging task that is currently almost exclusively accomplished by fluorescence in situ hybridization (FISH) techniques using probes to target different taxonomic groups (102–106), or extended to specific gene expression patterns in a spatial context (107). Despite its advantages, the application of FISH is limited in that the cells are inactivated through fixation and that probes are not species-specific. Together, these limitations complicate the interpretation of positive FISH-hybridization signals to corresponding functional activity. The use of genetically tagged fluorescently labeled focal strains is of an advantage here, as it can point to their live-specific spatial localization within the habitat and amidst resident strains. This approach can also be combined with suitable host models and live microscopy to obtain time-resolved information (108–110). Reduced complexity in the target microbiota and standardized habitats that enable non-invasive and live visualization would enormously simplify the characterization of the colonization process of a focal strain and could thus provide crucial dynamic information that is hard to obtain otherwise. This may be achieved, for example, by using microfluidic culturing systems, which enable control over spatial geometry, hydrodynamic and/or substrate flow, material properties, nutrient sources, or gradients of electron acceptors. In addition, microfluidics approaches present opportunities to study bacterial cell adhesion, invasion, and virulence in habitats that mimic those of the host (111). Bead-encapsulation (51) or picoliter droplet culturing systems can be used to propagate microcolonies or micro-assemblages of different strains that reflect discontinuous growth environments (42). This might be coupled with sequencing strategies to detect specific compositional biases between inoculants and resident strains.
TOOLS AND STRATEGIES FOR N+1 AND N−1 ENGINEERING
Learning from nature
Despite its conceptual simplicity, successfully achieving N+1/N−1 states is a major challenge. The main reason is that the outcomes of focal strain inoculations are governed by complex ecosystem processes (which iterative N+1/N−1 cycles themselves aim to uncover). To better understand these processes, it is helpful to focus our attention briefly on what is generally known about the growth and development of microbiomes. More particularly, it is important to understand the characteristics of dispersal processes and how they can inform rational N+1/N−1 engineering efforts.
Microbiomes are inherently dynamic entities subject to processes that change their composition and functional gene repertoires. Here, we collectively refer to these as microbiome “developmental” processes. The development of microbiomes is, first of all, dependent on the microbial taxa that they encompass and their functionalities. Microbiomes are connected to the rest of the natural environment, and therefore, any microbiota is at any time subject to dispersal processes that lead to inflow of new strains (from “outside” the microbiome) and outflow (loss) of resident strains due to migration or cell death (Fig. 4A). For example, a growing plant will become colonized by microbes from the soil microbiome, an insect eating from a plant leaf will ingest microbes from the phyllosphere microbiome, and the microbes in its droppings (the insect gut microbiome) may find their way back into the soil. Existing resident and newly incoming strains undergo growth and decline as a function of selective pressures, available nutrients, or changes in the habitat, leading to fluctuations in taxon abundances, extinctions, and an eventual natural homeostatic state for the microbiome. Processes within the microbiome also contribute to its diversification, due, for example, to mutational drift, phage infection or predation pressure, and horizontal gene transfer processes, which can cause the sudden outgrowth or decline of resident strains (Fig. 4A) (15, 112–114).
Fig 4.
Concepts of microbiome development and N+1/N−1 engineering. (A) Dispersion processes leading to in- and outflow of strains into the habitats that form the microbiome boundaries. The initial establishment may be dependent on active seeding or transmission of specific microbial strains into the habitat, followed by the passive inflow of further strains, some of which may invade the established microbiota. Priority, selective, and random processes further determine the development of the microbiome. (B) Community development as a succession of different individual strain populations (colored lines), selected by the local environmental conditions, priorities, inflow, mutation, and available nutrients. Modification of habitat properties occurs as a consequence of growth, interspecific interactions, and emergent microbiome system-level properties. (C) Examples of N+1 interventions to restore microbiota-homeostasis: in case of a dysbiosed microbiome resulting from pathogen invasion or in case of a pollutant-stressed microbiome. Lines indicate N+1 population development and general microbiota richness. Gradients point to (reduced) pollutant levels resulting from inoculant activity. (D) Creating nutritional niches for N+1 proliferation, for example, by selective inhibition of a resident niche occupant (cells with red crosses).
Dispersal of strains can be considered as natural (albeit uncontrolled) N+1/N−1 events, most of which may go unnoticed or do not lead to strain establishment (though they may lead to the replacement of strains with others with very similar properties). For example, in the human gut, various strains of Escherichia coli succeed one another at relatively short time intervals (115). Progressive strain replacement has also been observed for plant-interacting pseudomonads, which evolve over time with their plant host, leading to the disappearance of ancestor strains (116, 117). Wastewater treatment systems, which act as open pools of inflowing material, collect a wide variety of microbial taxa with a specific set of recurrent core members (118). In some instances, however, incoming strains are able to expand their population at the expense of the stability of the resident microbiome. In these instances, one could speak of strain invasion, potentially leading to long-term deleterious consequences for the homeostatic processes of the natural microbiome (119). Pathogens are best known for their invasive behavior (Fig. 4B). For example, gut pathogens such as Salmonella enterica serovar Typhimurium (S. Tm) have evolved to exploit and release niches for their own establishment (36). Extreme distortion and collapse of microbiome homeostasis can also originate from within the existing microbiota by opportunistic strains. For example, several endogenous pathogens (such as Clostridiodes difficile) can occur at relatively low abundances in an undisturbed gut microbiome, but their outgrowth can arise when the composition, size, and function of the resident population are significantly impacted as a result of antibiotic treatment (120).
Natural microbiomes strongly differ in their propensity, frequency, or range for dispersal, and individual strains likewise differ in their capability to establish in a new habitat, which has been associated to their growth competitiveness under available resources (121–123). Microbiomes existing in (more or less) open environments, such as soils, wastewater, or aquatic systems, as well as on skin, and plant leaves or roots, would intuitively be expected to be more strongly influenced by species dispersal (from other environments) than microbiomes in more closed environments, such as human, animal, or insect guts. Dispersal success and environmental filtering are further dependent on the occupation state of the habitat (124). Some habitats like guts or skin tissues of newborns and growing roots are rather pristine at the time of microbiota colonization and settlement (77, 113), whereas others may be fully occupied with microorganisms up to the system’s carrying capacity. Intuitively, a pristine habitat may be easier to colonize than a system at carrying capacity, for which colonization resistance (i.e., the occupation of most available niches) needs to be overcome. Colonization processes of pristine habitats have been studied in detail and are typically characterized by waves of succession of different taxa and turnover until some form of steady-state or homeostasis is reached (45). For example, during human development, there is a pronounced rapid strain evolution and turnover of the microbiota in early childhood, when the human colonic microbiome follows a succession as the taxa gradually occupy the available niches (77). Colonic succession has been suggested to be strongly deterministic within the habitat conditions (45) but is further dependent on environmental cues. This has been attested by the permanent changes in gut microbiota composition in children with chronic undernutrition (125) or high-fat diets (126).
Modes of strain dispersal also play a role in successful establishment. Dispersal can take the form of active microbial motility, like gliding on surfaces, swarming, or swimming towards chemicals (110, 127) or light (128, 129), or can be a passive process. For example, bacteria can passively disperse via transport by fungal hyphae in soil (130, 131), by wind or water flow, association to particles, food uptake, or macroorganism activity (124, 132). Active motility typically allows for dispersal at smaller spatial scales, while passive transport allows for dispersal at larger spatial scales. Successful dispersal also entails overcoming ecological bottlenecks, which may determine the founder population size necessary for colonization. Finally, some dispersal mechanisms may have been selected naturally to ensure successful (or controlled) invasion and colonization of habitats, such as gut microbiota transmission through fecal pellets (133), maternal brood balls (134, 135), or breastfeeding (113).
In summary, N+1/N−1 intervention methods can thus take inspiration from these different processes to guide, for example, the design of specific requirements for windows of inoculation opportunity, niche availability, transmission, and temporal or long-term establishment.
Considerations for N+1/N−1 implementation
The processes underlying the outcomes of artificially introducing or eliminating strains from a microbiome will be similar to natural processes taking place during strain dispersal or microbiota transmission, except that by the engineering effort known limitations can be overcome (e.g., founder cell population size or niche availability). As a consequence, one can expect that the growth or demise of a focal strain in a target habitat will similarly depend on multiple factors, such as the state (composition, spatial structure, and functionalities) of the resident microbiota, the dynamic time window for inoculation opportunities (priority effects), physico-chemical parameters of the habitat, niche availability for the focal strain, selection for its metabolic capacities within the context of the habitat, its sensitivity to predation, and any specific competitive traits that a focal strain may deploy (136). To target a state in which N+1 focal strains can proliferate and survive within the target microbiome, it is necessary that appropriate nutrients are available to generate the metabolic energy necessary for their growth and that they can tolerate or adapt to any biological or physico-chemical stresses present in that habitat. These considerations may lead to different approaches depending on the microbiome. Targeted colonization of the relatively pristine guts or skin tissues of newborns, or growing roots of fresh seedlings, will require different strategies than inoculation for restoration of disrupted microbiomes resulting from medical interventions or pathogen attacks or for reduction of pollutant loads in contaminated environments. Since N+1/N−1 strategies may be particularly envisioned in contexts of disrupted or dysbiotic microbiomes (Fig. 1C), nutrient availabilities and potentially unfavorable habitat conditions may play a crucial role in successful inoculant establishment and activity.
For example, one might aim to revert the consequences of an invading gut pathogen and the accompanying microbiome changes by the intake of a non-pathogenic niche competitor (37). This competitor would have to be able to proliferate in the dramatically altered gut nutrient conditions (137), in order to exclude the pathogen from its occupied niche (127) (Fig. 4C). As a different example, environmental changes imposed by diet shifts, fertilizers, antimicrobials, pesticides, or pollutants can disrupt microbiota composition and function, thereby altering the nutrient availability while imposing additional toxic stress. The goal of a focal strain here might be to reduce the toxic load, but to do so it must be able to take advantage of an excess of available nutrients to grow to a sufficiently large population to have an effect (Fig. 4C). Environmental toxicity can mask competition for resources and promotes facilitation between community members, which could help a focal strain establish (138).
In some cases, the intended outcome of an N+1 engineering strategy may be to achieve permanent establishment (“engraftment”) of a focal strain within a microbiome, whereas in others the aim may rather be its transient and reversible presence. Examples of the latter may include the degradation of a toxic compound within a polluted system or the elimination of a plant pathogen during the vulnerable growth period of a crop (Fig. 1C). Alternatively, intended N+1 states may consist of engrafting specific genetic properties into the resident community members by means of the focal strain, but without necessarily maintaining it in the microbiota. This strategy has been followed, for example, to transfer pollutant degradation properties on mobile genetic elements into resident community members (139), but also to specifically inhibit functions in a targeted host within a microbiota (140, 141) or induce its killing (142). Temporal and controlled maintenance of focal strains has been a major goal of bioengineering efforts, which may involve genetically constructing or selecting strains with auxotrophies or inducible killing mechanisms that could restrict the survival of the focal strain outside the targeted habitat (142–145).
N+1/N−1 engineering tools
A number of tools are available to carry out an N+1/N−1 intervention process. In the easiest conception, N+1 engineering entails inoculating (i.e., mixing, injecting, swallowing, transplanting, etc.) a focal strain into a resident community at a defined dosage of live cells (Fig. 4D). Strain inoculation may be accompanied by a provision of unique carbon substrates or growth factors (synbiotics) exclusive for the focal strain that are added to the habitat or host, with the expectation that such substrates may at least temporarily favor its proliferation within the new habitat (146). Some studies have considered priming the focal strain before its inoculation, such that its adaptation period is minimal and does not hinder its proliferation within the target microbiome (147).
N−1 engineering, conversely, aims to deliberately eliminate, reduce, or inactivate a focal strain within a microbiome. Several means are conceivable for achieving this state, some of which rely on N+1 to achieve N−1 (in this context, introducing a focal strain to remove another, Fig. 4C). One approach, for example, has been used to eliminate a pathogenic strain by introducing a niche competitor at large numbers, which colonizes the microbiome and accelerates the clearance of the pathogen (143). Competitive niche exclusion is likely the reason underlying the frequent natural appearance of strain successions in the gut, which could thus be turned into a viable N−1 procedure (120). Similarly, the introduction of naturally occurring or engineered auxotrophic variants could drive the targeted extinction of a focal strain (148, 149). A recent success in the elimination of a recurring human gut pathogen, C. difficile, was obtained by re-inoculation not by a single focal strains but by complex microbiota retrieved from the distal gastrointestinal tract of healthy donors, a principle now known as fecal transplantation (150). Remarkably, this leads to the replacement of some 80% of strains present in the gastrointestinal tract before the transplantation and stable engraftment (more than 5 years) of almost two-thirds of the introduced donor taxa (151).
A second approach relies on eliciting an infection of the N−1 focal strain with a bacterial virus such as a lytic bacteriophage (phage), which can lead to its death. Phage infection is highly strain-specific and can be “self-propagating,” in that infected and lysed target cells produce more phage particles that can then specifically infect neighboring cells (152). As a result of its selectivity, phage therapy could “spare” most of the members in a microbiome in contrast to broad-spectrum antibiotics (153). Phage killing of the focal strain could also be used to “open” the niche for colonization with non-pathogenic competitor strains of the same species, which are resistant to the applied phage. However, resistance to applied phages has been observed to rapidly arise and a combination therapy with antibiotics may, therefore, be necessary for effectively treating an infection (154). Phages are also considered as tools to restore dysbiosed microbiomes, in an approach termed phage rehabilitation (155, 156). As an alternative to infective phage particles, microbiomes may be dosed with purified phage proteins, such as endolysins, to target strain-specific lysis and removal (157, 158). Finally, purified phage tail-like particles known as tailocins are also being considered as tools to control or provoke subtle strain abundance changes within microbiomes (159).
Similarly, N−1 focal strains may be targeted by toxic proteins leading to their cell death. A proof of principle for this concept has been demonstrated, in which a donor strain transmitted a genetic construct into an (E. coli or Vibrio cholerae) focal strain by horizontal gene transfer, leading to the expression of a toxin that inhibited further cell division (142). In another example, E. coli engineered to produce a narrow-spectrum siderophore-bacteriocin Microcin MccI47 was introduced into the mouse gut as a live biotherapeutic agent, which was able to selectively inhibit Enterobacteriaceae (160, 161). Lastly, N−1 approaches could also rely on purified bacteriocins with high host specificity (162, 163), or on drugs, especially if they display a narrow host spectrum (13, 164, 165). However, even narrow-spectrum agents may still be affecting commensal bacterial strains and, therefore, secondary effects on resident microbiota should be an integral part of N+1/N−1 studies.
Niche engineering
Apart from the biological tools needed for N+1/N−1 engineering, one should also consider the niches that are necessary for the focal strain to proliferate. Engineering the niches themselves can then be used as a tool to restore, implement, or prevent the growth of focal strains (or to combine both, Fig. 1C and 4D). For example, a wastewater treatment plant may in a sense be considered a (very rough) niche-engineered system to facilitate specific biogeochemical processes such as carbon removal, nitrogen transformation, or phosphate uptake, by controlling oxygen inflow and carbon dosage. Microbiome niche precision engineering has, until now, not been very well-developed, apart from a few examples including the synbiotic additions to inoculated strains mentioned above (146). For precise niche engineering, one would first have to map the available functional and spatial niches in a microbiome—a first approximation of which could be achieved by genome-scale metabolic models from individual isolates or metagenomes (Fig. 5A). In addition, one would also have to get an idea of the niche “size,” i.e., the carrying capacity of the niche for functionally similar strains. As an example, most soils have a relative abundance of around 25% Proteobacteria, which may constitute hundreds of individual strains (166). At a typical bacterial cell density of 109 per gram of soil, it may thus be challenging to expect maintenance of an N+1 focal strain at much higher than 107 cells per gram. The true accessibility of a niche may also be dependent on some window of inoculation opportunity. For example, an N+1 focal strain co-inoculated with resident community members simultaneously (so that all members need to grow) may find a larger niche than when inoculated into a steady-state community (essentially no net growth of any of the community members). Temporarily liberating niches to allow recolonization by others may, therefore, be a strategy to accompany N+1 /N−1 interventions (167) (Fig. 4D). This can be achieved by using abiotic compounds (antimicrobials, food additives, diet shifts, pesticides, or others) that target certain members of the community (37, 168, 169). When applied non-recurrently, these compounds can create a transient niche for the focal strain that can be occupied (37). In the case of mammalian-associated communities, the adaptive immune system can be harnessed via targeted vaccination approaches. While immunotherapies are intuitively an N−1 approach, recent evidence indicates some antibody responses can enhance microbial fitness, making it a feasible, targeted approach for N+ 1 interventions (108, 170–175).
Fig 5.
Computational tools for N+1/N−1 engineering. (A) Genome-scale metabolic networks incorporate all known biochemistry of an organism based on functional annotation from individual genomes or binned metagenome sequence data and can be contextualized by integrating additional constraints and data. (B) Metabolic interaction maps for the N+1/N−1 focal strains can be reconstructed by graph-based or constraint-based methods. (C) Microbial community function and development can be simulated at different scales to predict the effects of N+1/N−1 focal strain inoculations.
N+1/N−1 computational tools
Computational models are powerful tools to better understand the inherent complexity of single species’ metabolism and the emerging metabolic capabilities in the context of a microbiome (Fig. 5). These methods can be used as platforms to integrate and analyze data from the experimental designs outlined above, as well as to generate hypotheses and guide the design of experimental setups. Functional genome annotation based on sequence similarity has enabled the reconstruction of metabolic networks representative of all known biotransformation capabilities of an organism (Fig. 5A). These networks can be translated into mathematical models, which are known as genome-scale models (GEMs). GEMs can be coupled with constraint-based optimization methods to allow the estimation of feasible flux distribution profiles that support an observed phenotype (e.g., growth) and help gain mechanistic understanding (176). Additional layers of information can be added to GEMs, including but not limited to thermodynamic constraints (177), gene expression (178, 179), regulation (180, 181), kinetic rate law expressions (182), and various types of omics data, to make GEMs context- and application-specific. In the context of multispecies microbiota, reduced GEMs (183, 184) that preserve network properties can be considered to increase computational efficiency.
The information contained in metabolic networks and GEMs of single organisms can be leveraged to infer possible outcomes of N+1/N−1 interventions through the prediction of metabolic interactions between different species. Qualitative predictions of interspecific metabolic interactions can be made using metabolic networks and graph-based approaches (Fig. 5B). A metabolic network can be represented as a graph where each node represents a metabolite and each edge a reaction. Efficient graph search algorithms can then be employed to identify alternative pathways from a source metabolite in species A to a target metabolite in species B (185). The existence and multiplicity of such pathways can hint at possible cooperation among the species and can be informative of the resilience of a community to perturbations. Similarly, this approach can be employed to examine how these pathways are affected after the addition or removal of a focal strain. For example, one would expect that taxa with high connectivity act as metabolic hubs, whose elimination can radically affect community composition and function. Finally, community networks can be enriched with hypothetical biochemistry based on the enzymatic functions present in each genome (186, 187), allowing the discovery of yet uncharacterized community functions.
Constraint-based optimization methods can be used to infer metabolic interactions in a community (Fig. 5B). To do this, several tools have been developed under assumptions tailored to different types of interactions (e.g., positive or negative interactions, pairwise, or higher-order interactions) (188–190). These methods generally involve two steps. First, the external nutrients available to the community are specified. Such external nutrients are provided either by the metabolism of the host or by the environment. In the second step, the metabolic interactions are reconstructed given the external nutrients and organisms’ metabolic capabilities, i.e., their uptake and secretion reactions. This process yields valuable information on how abiotic (nutrient availability) or biotic perturbations (adding or removing a member) can alter the interaction networks. In addition, the generation of alternative interaction profiles helps to account for the uncertainty and variability of microbiome composition.
Simulation and design of microbial communities
Like a population of a single taxon in a closed environment, a microbial community in a habitat can be assumed to grow and converge to some stable state [arguably subject to the same dispersal and turnover mechanisms as described above (191, 192)]. Simulating the microbiome’s composition and relative taxon abundances, and its global metabolic functioning, is a longstanding challenge. The least complex form of computational methods simulates microbial communities at a steady state without making any assumptions about the history or taxonomic successions in the community (Fig. 5C). Such methods allow for a variety of analyses, including capturing nutrient uptake and byproduct secretion (193), estimating internal metabolic fluxes (194, 195), predicting relative abundances (196), studying resource allocation to macromolecule synthesis (197), and designing synthetic communities with desired properties (198, 199). Similarly, such computational tools can be used to study how the addition or elimination of an existing member might cause the community to converge to a different steady state. Perturbations in community composition, i.e., biotic perturbations, can affect flux profiles, abundances, nutrient requirements, byproduct formation, or resource allocation.
The scope of computational simulations can then be extended by incorporating temporal species and habitat dynamics (Fig. 5C). Dynamic species models include two classes: ecological models and evolutionary models (200). Lotka-Volterra models are a classic example of the former. They include a phenomenological description of the system in the form of ordinary differential equations (201). Evolutionary models, on the other hand, study the evolution and variation of a genotype of species over long-time scales (200). Dynamic simulations based on GEMs can capture temporal variations in microbial abundances, extracellular metabolite concentrations, and macromolecule synthesis (179, 202–204). Such methods allow studying and predicting the effect of adding/removing a member as a function of time, where the simulations can be contextualized based on the initial conditions, growth properties, and species interactions (205).
To accelerate N+1/N−1 research efforts, another layer of complexity is needed, which accounts for the spatiotemporal dynamics of microbial communities (Fig. 5C). Both synthetic and natural microbiota have an inherent spatial component that significantly affects their evolution and function. Microbial cells take up diffusing nutrients from their environment to grow and divide, driving the community to expand into spatial structures such as biofilms or cellular aggregates. The niche architecture is, therefore, constantly changing due to the local variations in the concentrations of metabolites and the abundance of microbial cells. In addition, the close proximity of microbial cells creates a crowded environment and reduces diffusion, which affects nutrient availability and the metabolic capabilities of the organisms (206). Spatiotemporal modeling approaches can simulate such complex biophysical (crowded) cell environments and can help us understand the stochastic effects of the N+1/N−1 events that are not captured by other modeling frameworks. These approaches differ in the level of detail they capture, often involving a trade-off between spatial resolution and computational cost (201). These models can incorporate the various biophysical and mechanical properties of the systems, such as cell shoving (207, 208), production of extracellular polymeric substances (209, 210), crowding effects (206, 211), cell morphology effects (212), complex habitat geometries (213, 214), and (flow) hydrodynamics (209, 215). Spatially resolved modeling of the cell environment and cell-cell interactions can help to understand how perturbations in abiotic or biotic factors (N+1/N−1) affect the system’s emergent properties (201, 216). Finally, such models help to predict changes in microbial interactions and functions between well-mixed and spatially organized environments.
N+1/N−1 INTERVENTION APPLICATIONS
Plant-beneficial bacteria and/or arbuscular mycorrhizal fungi in the rhizosphere
Plant-beneficial bacterial inoculants in agriculture have long been studied as substitutes for chemical fertilizers and pesticides, being able to ward off phytopathogens or insect pests, or to provide further beneficial functions such as solubilization of nutrients or priming for plant defense and stress tolerance (26, 217, 218). In practice, however, plant-beneficial strains typically only transiently establish, e.g., within the rhizosphere microbiome (219). Hence, multiple inoculations are needed, leading to temporary population increases followed by progressive decline until the next inoculation (27). The reasons that underlie this transient rhizosphere colonization are multiple and intricate. The inoculant must persist and be active in order to be able to exert its plant-beneficial functions in the rhizosphere. This implies that the inoculant must adapt to the particular physico-chemical and biotic conditions in the rhizosphere environment, including varying soil pH (220), water availability (221), or micro- and macronutrient accessibility (222, 223), but also to plant immune responses that can involve the release of signaling compounds (224). Root exudates provide carbon-rich compounds at the root surface and soil interface, which can support bacterial growth at high numbers (225). Nonetheless, plant roots also secrete secondary metabolites such as coumarins, flavones, and benzoxazinoids that can promote or inhibit the growth of specific taxa (226–229). Bacterial inoculants, in turn, may release metabolites that allow them to shape the root microenvironment (230–233), as well as modify plant metabolism and defense (234–236). These processes then indirectly alter the root microbiota composition.
As discussed in general terms above, the success of a beneficial inoculant to invade the rhizosphere microbiome is dependent on niche competition for resources or space. This competition is often mediated by broad-spectrum diffusible antimicrobial compounds, such as phenazines or acylphloroglucinols (237), and by specialized weaponry, such as contact-dependent type VI secretion systems (238), bacteriocins (239), or tailocins (240, 241). In addition, the survival of the plant-beneficial inoculant can be affected by natural predators inhabiting the rhizosphere microbiome, such as bacteriophages (242, 243) or grazing phagotrophic protists (244). The establishment of a newcomer into an otherwise settled rhizosphere microbiome could further influence the microbiome composition by altering the structure of key microbial functional groups, such as the nitrogen-fixing bacterial community (245). These modifications can ultimately have repercussions on plant growth and development (245–247).
Agricultural practice could also benefit from the selective removal of undesired taxa, notably bacterial, fungal, or oomycete phytopathogens that hinder plant development, impair plant health, and reduce crop yields (217, 248, 249). This is the long-desired goal of biocontrol strategies aimed at removing plant pathogens. Most biocontrol techniques have focused on introducing specific plant-beneficial strains, which can directly compete for resources or space with the pathogen, causing niche exclusion (26, 237). However, there are also approaches that would potentially allow the selective removal of an undesired member of the rhizosphere or soil microbial community without the introduction of another individual. For example, bacteriophages that directly prey on specific bacterial plant pathogens such as Ralstonia or Pectobacterium species were introduced as seed, tuber, or soil treatments (250). Similarly, the direct application of taxon-specific bacteriocins or tailocins to leaves resulted in diminished populations of pathogenic Pseudomonas and Xanthomonas and reduced disease incidence (251–253).
Future strategies for successful introduction of plant-beneficial inoculants should implement interactions between the inoculant and the plant microbiota in genotype-specific plant breeding strategies and foster agricultural measures such as soil amendments or adapted cropping sequences aimed at sustaining inoculant establishment and function (247, 254, 255). Many studies where bacteria are added to field soils have focused on the effects of inoculation (e.g., on plant growth or protection). However, few have followed how successful the added bacterial inoculum was in colonizing the soil or rhizosphere or have even examined the factors that determine whether or not it was successful.
Bioremediation and bioaugmentation in soil and wastewater systems
The concept of inoculating individual suspensions or mixtures of pure culture strains to induce or augment targeted metabolic processes within a microbiome (hence the term bioaugmentation) has been intensively and long practiced in the areas of soil and wastewater bioremediation (29, 256–258). Here, the underlying premise is that strains that have naturally adapted or evolved to use specific pollutants as carbon, nitrogen, or energy sources can be purified, isolated, and transplanted from one (contaminated) environment into another (259). Furthermore, specific cases of engineered bacteria with additional properties to their metabolic capacities, such as to better withstand toxicity, have been considered for bioaugmentation (22, 260–262). Strain inoculation has mostly been a trial-and-error approach (263–267), with emphasis on the outcomes of pollutant disappearance and bioavailability (263, 268–270), but little on understanding the underlying reasons or mechanisms for these outcomes. Fundamental questions as to whether strain characteristics predetermine success or whether there is a need for specific native ecological networks (given that strains do not come from the same site as where they are introduced) have traditionally not received the necessary attention (257). Only more recently have studies emerged that combine systematic efforts to determine inoculant properties or genetic factors that may support or disfavor their (temporary) establishment, to characterize the processes taking place during inoculation, survival, or strain demise, to unveil any commonalities among strains that may prime them for being effective colonizers, and to simultaneously uncover properties and reactions within the targeted microbiome [e.g., references (60, 61, 271, 272)].
Several studies using different inoculants intended for the degradation of aromatic compounds, for instance, have shown that physiological adaptations to a contaminated soil environment typically involve upregulation of stress defense systems, heavy metal resistance, nutrient scavenging, adhesion factors, and downregulation of motility (147, 273–277). Importantly, such adaptations are within the scope of the focal strains’ inherent properties, leading to their growth and temporal establishment within the target microbiome (60, 272), which is to a large extent dependent on the bioavailability of the targeted compound (147, 277, 278). As it can be assumed that the presence of the pollutant for which the N+1 inoculant is intended also presents its specific colonizable nutrient niche, the proliferation of the inoculant is favored for as long as the compound is available, and no other factors limit its activity or lead to its decline. Though it is not straightforward to understand at which point inherent strain properties break down and result in the strain’s decline, some studies have pointed to trivial yet difficult-to-overcome biological factors such as predation (256, 279, 280).
Unsurprisingly, studies using omics approaches have found evidence for the importance of a multitude of microbial genes for survival and proliferation in natural colonized habitats (60, 278). Their interpretation is complicated by the many unknown and hypothetical gene functions that appear to give selective benefit for growth and that point to multiple different mechanisms that enable N+1-inoculants to proliferate (273, 281). Obviously, contaminated soil is a very heterogeneous environment in which additional nutrients are limiting, which potentially hinders the population development of N+1 inoculants. Specific advantages for their proliferation may be found in spatially localized substrate provision from plant roots, leading to rhizo- (282, 283) and phytoremediation efforts (30, 281).
Food and nutrition
Probiotics are live microorganisms that, when administered in adequate amounts, may confer a health benefit on the host (284). The use of probiotics in the form of freeze-dried or liquid products, or cultured as part of food fermentation, is pervasive in modern nutrition and has a long tradition. This application has more recently been expanded with specific probiotic strains or strain mixtures, whose use has been claimed to result in potential benefits against obesity (285, 286), for stimulation of the immune system (287), or (re-)development of a homeostatic gut microbiota (113, 288–290). One could, therefore, consider the use of probiotics as an example of an N+1 intervention in the human gut microbiome. However, as in the two other application areas mentioned above, there is a strong focus concerned with clinical or health outcomes, whereas probiotic colonization itself remains complex and controversial (290). In addition, understanding the mechanisms of action, causalities, and impacts of probiotics on microbiome development is still limited and under debate.
Today, the most commonly used probiotics include Bifidobacterium and Lactobacillus strains (291), but also Streptococcus salivarius, Enterococcus faecium, Akkermansia muciniphila (286), or Anaerobutyricum soehngenii (292). Their application (in the sense of the N+1 concept) consists of high dosages (109–1010 live bacteria per intake), taken daily over months (287, 288). Available measurements of, e.g., stool content seem to indicate that the swallowed probiotics do not engraft within the (human) gut environment and need to sustain high constant titers to exert their action. For example, mothers in a mother-infant cohort ingested capsules with four probiotic strains (total 1.2 × 1010 colony forming units) daily for 6 months until birth, and then the newborns followed this pattern for 3 months. Significant probiotic strain survival (reaching up to 80% of relative abundances) was observed only when infants were breastfed, and mostly for Bifidobacterium breve (288). Pre- and probiotics, therefore, often accompany each other, with the purpose to provide specific carbon sources or other nutrients to the probiotic strain (146, 290, 293).
Despite demonstrated physiological effects on the host, there is also evidence that intake of probiotics may be harmful, particularly in sensitive groups or individuals (294). This risk highlights the need for a precise understanding of the mechanisms, causality, impact, and safety of probiotics (292). For example, the effects of Bifidobacterium longum administration may be dependent on the composition of the underlying gut microbiota and differences in intestinal cell wall adhesion (295). A more thorough and personalized understanding of the risks and benefits of these treatments may be achieved through targeted microbiota interventions in an N+1/N−1 framing.
Enteropathogenic infections in the animal gut
The composition and function of the microbiota in the animal gut are affected not only by available nutrients, interactions between its members, and physical constraints (size of the gut, gut transit times, body temperature, etc.) but also by the host immune defenses. The latter can specifically enhance the removal of unwanted microbes and the colonization efficiency of newcomers (296). Moreover, host genetic variation also contributes to microbial colonization efficiency (297).
The gut is a portal of infection by a significant number of food-borne enteropathogenic bacteria, which cause much morbidity and mortality worldwide (298). Of particular relevance are members of the Enterobacteriaceae, including pathogenic E. coli, Shigella, and Salmonella strains. These enteropathogens are ideal focal strains for N+1/N−1 studies, because of their high prevalence, their genetic accessibility, a vast body of knowledge on E. coli and S. enterica serovar Typhimurium growth physiology and metabolism, and a multitude of robust ex vivo and in vivo infection models [reviewed in references (168, 299, 300)]. Their study has greatly helped to understand the mechanisms promoting infection, which, as mentioned above, resembles a natural N+1 situation. In addition, studies on enteropathogenic infections have helped to understand the role of colonization resistance in preventing infectious diarrhea after pathogen ingestion. Finally, many of the effects of the gut microbiota on host immune responses during the course of an infection and frequent spontaneous pathogen elimination when the host recovers from acute diarrhea have been detailed and summarized (168, 296).
The resident gut microbiota normally provides resistance against colonization of a food-ingested pathogen such as S. Tm, limiting its growth in the gut lumen and thereby preventing gut tissue invasion and enteric disease in many infected hosts. This has been inferred from epidemiological data (301) and mouse experiments. Many experimental infection models rely on microbiome perturbation prior to pathogen introduction to establish colonization and pathogenesis (169, 302, 303). Complex gut microbiota indeed efficiently protect mice from Salmonella gut colonization, and the effect of colonization resistance can be increased by introducing competitive E. coli strains (37, 38, 304, 305). Recent work suggests that strain-specific capacities to utilize poly-arabinose fibers, arabinose, or sugar alcohols like galactitol can enhance their gut-luminal growth (306, 307). This might provide a concept that can be used to create niches that would select for proliferation of added competitive Enterobacteriaceae strains and could prevent or reduce S. Tm diarrhea and its spread in food animal herds.
S. Tm gut colonization is further suppressed by the host’s adaptive immune system, in particular, by secretory Immunoglobulin A, which crosslinks S. Tm daughter cells during division, leading to large aggregates. This prevents tissue invasion and favors more efficient clearing by the fecal stream (108). Moreover, recent work has further indicated that oral vaccination in combination with N+1 competitor E. coli strains can suppress S. Tm gut colonization (N−1) in otherwise susceptible mice (308). Finally, an intestinal colonization-competent auxotrophic S. Tm vaccine strain induced an immune response in mice and acted as a niche competitor (143). Such proof-of-concept studies in mice illustrate that combining orthogonal approaches can provide powerful tools or therapies to prevent infection via N+1/N−1 strategies.
OUTLOOK
The field of microbiome engineering is still in its infancy. Nonetheless, the wide variety of studies in a range of microbiome systems has already presented a wealth of information on potential strategies for microbiome interventions. What is clear from the overview presented here is that much can be learned from cross-microbiome efforts, coupled with general conceptual theory on natural microbiome development. While immediate success might be observed in specific applications related to potential health or environmental benefit, behavior and processes are often more complex than initially assumed. There is thus a strong need to start small: with single focal strains, including specific designs that aim to study strain behavior and survival in the context of the microbiome, and including the most appropriate approaches to understand strain and system function, as illustrated here. Such approaches could also embrace reintroduction attempts of strains that were depleted in the recent past (309), or that are geographically distinct (290).
Advancing general theory is difficult but will greatly benefit from the depth of data that can be achieved using omics techniques and analyses of clinical or environmental parameters. Crucial for understanding cross-system N+1/N−1 processes are computational approaches, consisting not only of the data analyses that underlie most of the experimental designs outlined above but also of specific computational efforts to simulate or compare N+1/N−1 inoculations with natural events. Ideally, one would be able to predict the growth and survival of focal strains within target microbiomes under the conditions of their native habitats. Considering that growth relies on energy generating and anabolic processes, much will be gained by the improvement and application of computational—and specifically genome-scale, models. The in silico behavior of an N+1 focal strain could thus be approached within its environmental context. Combining multiple individual genome-scale models into a coherent microbiome model is still a daunting task, but first efforts show how important metabolic features and fluxes between taxa can be approximated.
Finally, it is important to capture community assembly processes within the context of the habitat, as a function of physical processes, microbial activity, and taxonomic composition using computational frameworks. As discussed, a variety of approaches are available for this task, even for models that predict the compositional development of communities based on carbon, nutrient, and energy input or as a function of assumed (imposed) interspecific interactions. Models can further be discretized to allow the division of individual “agents” (virtual cells) of different taxa within defined space that add cell movement, and allow nutrient gradients, diffusion terms, or cell crowding effects. The agents can be subject to more general deterministic laws of growth physiology (e.g., Monod) or interspecific interactions (e.g., Lotka-Volterra), but can also be represented by reduced genome-scale models for increased computational efficiency while still maintaining a representative set of metabolic reactions. Harmonized strategies like the N+1/N− concept may thus eventually facilitate the comparison of ecological roles, organism characteristics, and effects of perturbations on a system scale across studies and microbiomes. Crucially, these advances will benefit greatly from appropriate data accessibility and stewardship practices in microbiome research (310–312).
ACKNOWLEDGMENTS
This work was supported by the Swiss National Centre for Competence in Research (NCCR) Microbiomes and is the outcome of many discussions among the various consortium members. W.D.H. acknowledges the support of Horizon 2020 Marie Skłodowska-Curie grant agreement No. 956279. A.R.P. is supported by a James. S. McDonnell Postdoctoral Fellowship (2020–1332). The funders had no role in study design, data collection, and interpretation, or the decision to submit the work for publication.
The authors are junior and senior members of the Swiss National Centre for Competence in Research (NCCR) Microbiomes, a collaborative network of 24 research groups with the goal of advancing our understanding of microbiome assembly, development, and functioning, across human, animal, plant, and environmental systems. In addition, the network aims to develop, test, and improve methodologies to intervene in malfunctioning microbiomes, or build synthetic microbiomes with specific functionalities. This article was written on the basis of circulating discussions and concepts from within the network, with authorship based on the voluntary participation of anyone in the consortium. More information on the NCCR Microbiomes can be found at https://nccr-microbiomes.ch.
This article is dedicated to the loving memory of Alma Dal Co.
The authors declare no financial conflicts of interest.
Contributor Information
Jan Roelof van der Meer, Email: Janroelof.vandermeer@unil.ch.
Michael G. Thomas, University of Wisconsin-Madison, Madison, Wisconsin, USA
REFERENCES
- 1. Berg G, Rybakova D, Fischer D, Cernava T, Vergès M-C, Charles T, Chen X, Cocolin L, Eversole K, Corral GH, Kazou M, Kinkel L, Lange L, Lima N, Loy A, Macklin JA, Maguin E, Mauchline T, McClure R, Mitter B, Ryan M, Sarand I, Smidt H, Schelkle B, Roume H, Kiran GS, Selvin J, de Souza RSC, van Overbeek L, Singh BK, Wagner M, Walsh A, Sessitsch A, Schloter M. 2020. Microbiome definition re-visited: old concepts and new challenges. Microbiome 8:119. doi: 10.1186/s40168-020-00905-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Fierer N. 2017. Embracing the unknown: disentangling the complexities of the soil microbiome. Nat Rev Microbiol 15:579–590. doi: 10.1038/nrmicro.2017.87 [DOI] [PubMed] [Google Scholar]
- 3. Müller DB, Vogel C, Bai Y, Vorholt JA. 2016. The plant microbiota: systems-level insights and perspectives. Annu Rev Genet 50:211–234. doi: 10.1146/annurev-genet-120215-034952 [DOI] [PubMed] [Google Scholar]
- 4. Gilbert JA, Quinn RA, Debelius J, Xu ZZ, Morton J, Garg N, Jansson JK, Dorrestein PC, Knight R. 2016. Microbiome-wide association studies link dynamic microbial consortia to disease. Nature 535:94–103. doi: 10.1038/nature18850 [DOI] [PubMed] [Google Scholar]
- 5. Kostic AD, Howitt MR, Garrett WS. 2013. Exploring host-microbiota interactions in animal models and humans. Genes Dev 27:701–718. doi: 10.1101/gad.212522.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Raymann K, Moran NA. 2018. The role of the gut microbiome in health and disease of adult honey bee workers. Curr Opin Insect Sci 26:97–104. doi: 10.1016/j.cois.2018.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Tropini C, Earle KA, Huang KC, Sonnenburg JL. 2017. The gut microbiome: connecting spatial organization to function. Cell Host Microbe 21:433–442. doi: 10.1016/j.chom.2017.03.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Engel P, Moran NA. 2013. The gut microbiota of insects - diversity in structure and function. FEMS Microbiol Rev 37:699–735. doi: 10.1111/1574-6976.12025 [DOI] [PubMed] [Google Scholar]
- 9. Shade A, Peter H, Allison SD, Baho DL, Berga M, Bürgmann H, Huber DH, Langenheder S, Lennon JT, Martiny JBH, Matulich KL, Schmidt TM, Handelsman J. 2012. Fundamentals of microbial community resistance and resilience. Front Microbiol 3:417. doi: 10.3389/fmicb.2012.00417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Allison SD, Martiny JBH. 2008. Colloquium paper: resistance, resilience, and redundancy in microbial communities. Proc Natl Acad Sci U S A 105 Suppl 1:11512–11519. doi: 10.1073/pnas.0801925105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Herold M, Martínez Arbas S, Narayanasamy S, Sheik AR, Kleine-Borgmann LAK, Lebrun LA, Kunath BJ, Roume H, Bessarab I, Williams RBH, Gillece JD, Schupp JM, Keim PS, Jäger C, Hoopmann MR, Moritz RL, Ye Y, Li S, Tang H, Heintz-Buschart A, May P, Muller EEL, Laczny CC, Wilmes P. 2020. Integration of time-series meta-omics data reveals how microbial ecosystems respond to disturbance. Nat Commun 11:5281. doi: 10.1038/s41467-020-19006-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Philippot L, Griffiths BS, Langenheder S. 2021. Microbial community resilience across ecosystems and multiple disturbances. Microbiol Mol Biol Rev 85:e00026–00020. doi: 10.1128/MMBR.00026-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, Patil KR, Bork P, Typas A. 2018. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature 555:623–628. doi: 10.1038/nature25979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Vorholt JA, Vogel C, Carlström CI, Müller DB. 2017. Establishing causality: opportunities of synthetic communities for plant microbiome research. Cell Host Microbe 22:142–155. doi: 10.1016/j.chom.2017.07.004 [DOI] [PubMed] [Google Scholar]
- 15. Vellend M. 2010. Conceptual synthesis in community ecology. Q Rev Biol 85:183–206. doi: 10.1086/652373 [DOI] [PubMed] [Google Scholar]
- 16. Hattich GSI, Listmann L, Govaert L, Pansch C, Reusch TBH, Matthiessen B. 2022. Experimentally decomposing phytoplankton community change into ecological and evolutionary contributions. Funct Ecol 36:120–132. doi: 10.1111/1365-2435.13923 [DOI] [Google Scholar]
- 17. Malard LA, Mod HK, Guex N, Broennimann O, Yashiro E, Lara E, Mitchell EAD, Niculita-Hirzel H, Guisan A. 2022. Comparative analysis of diversity and environmental niches of soil bacterial, archaeal, fungal and protist communities reveal niche divergences along environmental gradients in the Alps. Soil Biology and Biochemistry 169:108674. doi: 10.1016/j.soilbio.2022.108674 [DOI] [Google Scholar]
- 18. Paoli L, Ruscheweyh H-J, Forneris CC, Hubrich F, Kautsar S, Bhushan A, Lotti A, Clayssen Q, Salazar G, Milanese A, Carlström CI, Papadopoulou C, Gehrig D, Karasikov M, Mustafa H, Larralde M, Carroll LM, Sánchez P, Zayed AA, Cronin DR, Acinas SG, Bork P, Bowler C, Delmont TO, Gasol JM, Gossert AD, Kahles A, Sullivan MB, Wincker P, Zeller G, Robinson SL, Piel J, Sunagawa S. 2022. Biosynthetic potential of the global ocean microbiome. Nature 607:111–118. doi: 10.1038/s41586-022-04862-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Coelho LP, Alves R, Del Río ÁR, Myers PN, Cantalapiedra CP, Giner-Lamia J, Schmidt TS, Mende DR, Orakov A, Letunic I, Hildebrand F, Van Rossum T, Forslund SK, Khedkar S, Maistrenko OM, Pan S, Jia L, Ferretti P, Sunagawa S, Zhao X-M, Nielsen HB, Huerta-Cepas J, Bork P. 2022. Towards the biogeography of prokaryotic genes. Nature 601:252–256. doi: 10.1038/s41586-021-04233-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Lindemann SR, Bernstein HC, Song H-S, Fredrickson JK, Fields MW, Shou W, Johnson DR, Beliaev AS. 2016. Engineering microbial consortia for controllable outputs. ISME J 10:2077–2084. doi: 10.1038/ismej.2016.26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Baker KF, Synder WC. 1965. Ecology of soil-borne plant pathogens—prelude to biological control. University of California Press, Berkeley. [Google Scholar]
- 22. Rojo F, Pieper DH, Engesser KH, Knackmuss HJ, Timmis KN. 1987. Assemblage of ortho cleavage route for simultaneous degradation of chloro- and methylaromatics. Science 238:1395–1398. doi: 10.1126/science.3479842 [DOI] [PubMed] [Google Scholar]
- 23. Verschuere L, Rombaut G, Sorgeloos P, Verstraete W. 2000. Probiotic bacteria as biological control agents in aquaculture. Microbiol Mol Biol Rev 64:655–671. doi: 10.1128/MMBR.64.4.655-671.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Saxelin M, Tynkkynen S, Mattila-Sandholm T, de Vos WM. 2005. Probiotic and other functional microbes: from markets to mechanisms. Curr Opin Biotechnol 16:204–211. doi: 10.1016/j.copbio.2005.02.003 [DOI] [PubMed] [Google Scholar]
- 25. Stecher B, Hardt WD. 2011. Mechanisms controlling pathogen colonization of the gut. Curr Opin Microbiol 14:82–91. doi: 10.1016/j.mib.2010.10.003 [DOI] [PubMed] [Google Scholar]
- 26. Haas D, Défago G. 2005. Biological control of soil-borne pathogens by fluorescent pseudomonads. Nat Rev Microbiol 3:307–319. doi: 10.1038/nrmicro1129 [DOI] [PubMed] [Google Scholar]
- 27. Tabassum B, Khan A, Tariq M, Ramzan M, Iqbal Khan MS, Shahid N, Aaliya K. 2017. Bottlenecks in commercialisation and future prospects of PGPR. Appl Soil Ecol 121:102–117. doi: 10.1016/j.apsoil.2017.09.030 [DOI] [Google Scholar]
- 28. Endt K, Stecher B, Chaffron S, Slack E, Tchitchek N, Benecke A, Van Maele L, Sirard J-C, Mueller AJ, Heikenwalder M, Macpherson AJ, Strugnell R, von Mering C, Hardt W-D, Stebbins CE. 2010. The microbiota mediates pathogen clearance from the gut lumen after non-typhoidal Salmonella diarrhea. PLoS Pathog 6:e1001097. doi: 10.1371/journal.ppat.1001097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Cycoń M, Mrozik A, Piotrowska-Seget Z. 2017. Bioaugmentation as a strategy for the remediation of pesticide-polluted soil: a review. Chemosphere 172:52–71. doi: 10.1016/j.chemosphere.2016.12.129 [DOI] [PubMed] [Google Scholar]
- 30. Simmer RA, Schnoor JL. 2022. Phytoremediation, bioaugmentation, and the plant microbiome. Environ Sci Technol 56:16602–16610. doi: 10.1021/acs.est.2c05970 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Agler MT, Ruhe J, Kroll S, Morhenn C, Kim S-T, Weigel D, Kemen EM. 2016. Microbial hub taxa link host and abiotic factors to plant microbiome variation. PLoS Biol 14:e1002352. doi: 10.1371/journal.pbio.1002352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Herren CM, McMahon KD. 2018. Keystone taxa predict compositional change in microbial communities. Environ Microbiol 20:2207–2217. doi: 10.1111/1462-2920.14257 [DOI] [PubMed] [Google Scholar]
- 33. Rivett DW, Bell T. 2018. Abundance determines the functional role of bacterial phylotypes in complex communities. Nat Microbiol 3:767–772. doi: 10.1038/s41564-018-0180-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Schmidt F, Zimmermann J, Tanna T, Farouni R, Conway T, Macpherson AJ, Platt RJ. 2022. Noninvasive assessment of gut function using transcriptional recording sentinel cells. Science 376:eabm6038. doi: 10.1126/science.abm6038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Studer N, Desharnais L, Beutler M, Brugiroux S, Terrazos MA, Menin L, Schürch CM, McCoy KD, Kuehne SA, Minton NP, Stecher B, Bernier-Latmani R, Hapfelmeier S. 2016. Functional intestinal bile acid 7α-dehydroxylation by Clostridium scindens associated with protection from Clostridium difficile infection in a gnotobiotic mouse model. Front Cell Infect Microbiol 6:191. doi: 10.3389/fcimb.2016.00191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Nguyen BD, Cuenca V M, Hartl J, Gül E, Bauer R, Meile S, Rüthi J, Margot C, Heeb L, Besser F, Escriva PP, Fetz C, Furter M, Laganenka L, Keller P, Fuchs L, Christen M, Porwollik S, McClelland M, Vorholt JA, Sauer U, Sunagawa S, Christen B, Hardt W-D. 2020. Import of aspartate and malate by DcuABC drives H2/fumarate respiration to promote initial Salmonella gut-lumen colonization in mice. Cell Host Microbe 27:922–936. doi: 10.1016/j.chom.2020.04.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Wotzka SY, Kreuzer M, Maier L, Arnoldini M, Nguyen BD, Brachmann AO, Berthold DL, Zünd M, Hausmann A, Bakkeren E, Hoces D, Gül E, Beutler M, Dolowschiak T, Zimmermann M, Fuhrer T, Moor K, Sauer U, Typas A, Piel J, Diard M, Macpherson AJ, Stecher B, Sunagawa S, Slack E, Hardt WD. 2019. Escherichia coli limits Salmonella Typhimurium infections after diet shifts and fat-mediated microbiota perturbation in mice. Nat Microbiol 4:2164–2174. doi: 10.1038/s41564-019-0568-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Brugiroux S, Beutler M, Pfann C, Garzetti D, Ruscheweyh HJ, Ring D, Diehl M, Herp S, Lötscher Y, Hussain S, Bunk B, Pukall R, Huson DH, Münch PC, McHardy AC, McCoy KD, Macpherson AJ, Loy A, Clavel T, Berry D, Stecher B. 2016. Genome-guided design of a defined mouse microbiota that confers colonization resistance against Salmonella enterica serovar Typhimurium. Nat Microbiol 2:16215. doi: 10.1038/nmicrobiol.2016.215 [DOI] [PubMed] [Google Scholar]
- 39. Schäfer M, Vogel CM, Bortfeld-Miller M, Mittelviefhaus M, Vorholt JA. 2022. Mapping phyllosphere microbiota interactions in planta to establish genotype-phenotype relationships. Nat Microbiol 7:856–867. doi: 10.1038/s41564-022-01132-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Grosskopf T, Soyer OS. 2014. Synthetic microbial communities. Curr Opin Microbiol 18:72–77. doi: 10.1016/j.mib.2014.02.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Hsu RH, Clark RL, Tan JW, Ahn JC, Gupta S, Romero PA, Venturelli OS. 2019. Microbial interaction network inference in microfluidic droplets. Cell Syst 9:229–242. doi: 10.1016/j.cels.2019.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kehe J, Kulesa A, Ortiz A, Ackerman CM, Thakku SG, Sellers D, Kuehn S, Gore J, Friedman J, Blainey PC. 2019. Massively parallel screening of synthetic microbial communities. Proc Natl Acad Sci U S A 116:12804–12809. doi: 10.1073/pnas.1900102116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Celiker H, Gore J. 2014. Clustering in community structure across replicate ecosystems following a long-term bacterial evolution experiment. Nat Commun 5:4643. doi: 10.1038/ncomms5643 [DOI] [PubMed] [Google Scholar]
- 44. Bai Y, Müller DB, Srinivas G, Garrido-Oter R, Potthoff E, Rott M, Dombrowski N, Münch PC, Spaepen S, Remus-Emsermann M, Hüttel B, McHardy AC, Vorholt JA, Schulze-Lefert P. 2015. Functional overlap of the Arabidopsis leaf and root microbiota. Nature 528:364–369. doi: 10.1038/nature16192 [DOI] [PubMed] [Google Scholar]
- 45. Čaušević S, Tackmann J, Sentchilo V, von Mering C, van der Meer JR. 2022. Reproducible propagation of species-rich soil bacterial communities suggests robust underlying deterministic principles of community formation. mSystems 7:e0016022. doi: 10.1128/msystems.00160-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Kešnerová L, Mars RAT, Ellegaard KM, Troilo M, Sauer U, Engel P. 2017. Disentangling metabolic functions of bacteria in the honey bee gut. PLoS Biol 15:e2003467. doi: 10.1371/journal.pbio.2003467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Brochet S, Quinn A, Mars RAT, Neuschwander N, Sauer U, Engel P. 2021. Niche partitioning facilitates coexistence of closely related honey bee gut bacteria. Elife 10:e68583. doi: 10.7554/eLife.68583 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Afrizal A, Jennings SAV, Hitch TCA, Riedel T, Basic M, Panyot A, Treichel N, Hager FT, Wong EO-Y, Wolter B, Viehof A, von Strempel A, Eberl C, Buhl EM, Abt B, Bleich A, Tolba R, Blank LM, Navarre WW, Kiessling F, Horz H-P, Torow N, Cerovic V, Stecher B, Strowig T, Overmann J, Clavel T. 2022. Enhanced cultured diversity of the mouse gut microbiota enables custom-made synthetic communities. Cell Host Microbe 30:1630–1645. doi: 10.1016/j.chom.2022.09.011 [DOI] [PubMed] [Google Scholar]
- 49. Stecher B, Chaffron S, Käppeli R, Hapfelmeier S, Freedrich S, Weber TC, Kirundi J, Suar M, McCoy KD, von Mering C, Macpherson AJ, Hardt W-D. 2010. Like will to like: abundances of closely related species can predict susceptibility to intestinal colonization by pathogenic and commensal bacteria. PLoS Pathog 6:e1000711. doi: 10.1371/journal.ppat.1000711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Cheng AG, Ho P-Y, Aranda-Díaz A, Jain S, Yu FB, Meng X, Wang M, Iakiviak M, Nagashima K, Zhao A, Murugkar P, Patil A, Atabakhsh K, Weakley A, Yan J, Brumbaugh AR, Higginbottom S, Dimas A, Shiver AL, Deutschbauer A, Neff N, Sonnenburg JL, Huang KC, Fischbach MA. 2022. Design, construction, and in vivo augmentation of a complex gut microbiome. Cell 185:3617–3636. doi: 10.1016/j.cell.2022.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Dubey M, Hadadi N, Pelet S, Carraro N, Johnson DR, van der Meer JR. 2021. Environmental connectivity controls diversity in soil microbial communities. Commun Biol 4:492. doi: 10.1038/s42003-021-02023-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Roggo C, Picioreanu C, Richard X, Mazza C, van Lintel H, van der Meer JR. 2018. Quantitative chemical biosensing by bacterial chemotaxis in microfluidic chips. Environ Microbiol 20:241–258. doi: 10.1111/1462-2920.13982 [DOI] [PubMed] [Google Scholar]
- 53. Smriga S, Ciccarese D, Babbin AR. 2021. Denitrifying bacteria respond to and shape microscale gradients within particulate matrices. Commun Biol 4:570. doi: 10.1038/s42003-021-02102-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Dal Co A, van Vliet S, Kiviet DJ, Schlegel S, Ackermann M. 2020. Short-range interactions govern the dynamics and functions of microbial communities. Nat Ecol Evol 4:366–375. doi: 10.1038/s41559-019-1080-2 [DOI] [PubMed] [Google Scholar]
- 55. Goldschmidt F, Regoes RR, Johnson DR. 2017. Successive range expansion promotes diversity and accelerates evolution in spatially structured microbial populations. ISME J 11:2112–2123. doi: 10.1038/ismej.2017.76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Doan HK, Antequera-Gómez ML, Parikh AN, Leveau JHJ. 2020. Leaf surface topography contributes to the ability of Escherichia coli on leafy greens to resist removal by washing, escape disinfection with chlorine, and disperse through splash. Front Microbiol 11:1485. doi: 10.3389/fmicb.2020.01485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Collins J, Auchtung JM, Schaefer L, Eaton KA, Britton RA. 2015. Humanized microbiota mice as a model of recurrent Clostridium difficile disease. Microbiome 3:35. doi: 10.1186/s40168-015-0097-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Liberti J, Kay T, Quinn A, Kesner L, Frank ET, Cabirol A, Richardson TO, Engel P, Keller L. 2022. The gut microbiota affects the social network of honeybees. Nat Ecol Evol 6:1471–1479. doi: 10.1038/s41559-022-01840-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Carlström CI, Field CM, Bortfeld-Miller M, Müller B, Sunagawa S, Vorholt JA. 2019. Synthetic microbiota reveal priority effects and keystone strains in the Arabidopsis phyllosphere. Nat Ecol Evol 3:1445–1454. doi: 10.1038/s41559-019-0994-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Morales M, Sentchilo V, Carraro N, Causevic S, Vuarambon D, van der Meer JR, Hug LA, Charles T. 2023. Fitness-conditional genes for soil adaptation in the bioaugmentation agent Pseudomonas veronii 1YdBTEX2. mSystems 8:e0117422. doi: 10.1128/msystems.01174-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Morales M, Sentchilo V, Hadadi N, van der Meer JR. 2021. Genome-wide gene expression changes of Pseudomonas veronii 1YdBTEX2 during bioaugmentation in polluted soils. Environ Microbiome 16:8. doi: 10.1186/s40793-021-00378-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. de Bakker V, Liu X, Bravo AM, Veening J-W. 2022. CRISPRi-seq for genome-wide fitness quantification in bacteria. Nat Protoc 17:252–281. doi: 10.1038/s41596-021-00639-6 [DOI] [PubMed] [Google Scholar]
- 63. Leveau JHJ, Lindow SE. 2002. Bioreporters in microbial ecology. Curr Opin Microbiol 5:259–265. doi: 10.1016/s1369-5274(02)00321-1 [DOI] [PubMed] [Google Scholar]
- 64. Remus-Emsermann MNP, Leveau JHJ. 2010. Linking environmental heterogeneity and reproductive success at single-cell resolution. ISME J 4:215–222. doi: 10.1038/ismej.2009.110 [DOI] [PubMed] [Google Scholar]
- 65. van der Meer JR, Belkin S. 2010. Where microbiology meets microengineering: design and applications of reporter bacteria. Nat Rev Microbiol 8:511–522. doi: 10.1038/nrmicro2392 [DOI] [PubMed] [Google Scholar]
- 66. Gil D, Bouché JP. 1991. ColE1-type vectors with fully repressible replication. Gene 105:17–22. doi: 10.1016/0378-1119(91)90508-9 [DOI] [PubMed] [Google Scholar]
- 67. Myhrvold C, Kotula JW, Hicks WM, Conway NJ, Silver PA. 2015. A distributed cell division counter reveals growth dynamics in the gut microbiota. Nat Commun 6:10039. doi: 10.1038/ncomms10039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Starr EP, Shi S, Blazewicz SJ, Probst AJ, Herman DJ, Firestone MK, Banfield JF. 2018. Stable isotope informed genome-resolved metagenomics reveals that Saccharibacteria utilize microbially-processed plant-derived carbon. Microbiome 6:122. doi: 10.1186/s40168-018-0499-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Riglar DT, Richmond DL, Potvin-Trottier L, Verdegaal AA, Naydich AD, Bakshi S, Leoncini E, Lyon LG, Paulsson J, Silver PA. 2019. Bacterial variability in the mammalian gut captured by a single-cell synthetic oscillator. Nat Commun 10:4665. doi: 10.1038/s41467-019-12638-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Bakkeren E, Herter JA, Huisman JS, Steiger Y, Gül E, Newson JPM, Brachmann AO, Piel J, Regoes R, Bonhoeffer S, Diard M, Hardt W-D. 2021. Pathogen invasion-dependent tissue reservoirs and plasmid-encoded antibiotic degradation boost plasmid spread in the gut. Elife 10:e69744. doi: 10.7554/eLife.69744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Hoces D, Greter G, Arnoldini M, Stäubli ML, Moresi C, Sintsova A, Berent S, Kolinko I, Bansept F, Woller A, Häfliger J, Martens E, Hardt W-D, Sunagawa S, Loverdo C, Slack E. 2023. Fitness advantage of Bacteroides thetaiotaomicron capsular polysaccharide in the mouse gut depends on the resident microbiota. Elife 12:e81212. doi: 10.7554/eLife.81212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Coronado E, Valtat A, van der Meer JR. 2015. Sphingomonas wittichii RW1 gene reporters interrogating the dibenzofuran metabolic network highlight conditions for early successful development in contaminated microcosms. Environ Microbiol Rep 7:480–488. doi: 10.1111/1758-2229.12276 [DOI] [PubMed] [Google Scholar]
- 73. Ackermann M, Stecher B, Freed NE, Songhet P, Hardt WD, Doebeli M. 2008. Self-destructive cooperation mediated by phenotypic noise. Nature 454:987–990. doi: 10.1038/nature07067 [DOI] [PubMed] [Google Scholar]
- 74. Hapfelmeier S, Stecher B, Barthel M, Kremer M, Müller AJ, Heikenwalder M, Stallmach T, Hensel M, Pfeffer K, Akira S, Hardt WD. 2005. The Salmonella pathogenicity Island (SPI)-2 and SPI-1 type III secretion systems allow Salmonella serovar Typhimurium to trigger colitis via MyD88-dependent and MyD88-independent mechanisms. J Immunol 174:1675–1685. doi: 10.4049/jimmunol.174.3.1675 [DOI] [PubMed] [Google Scholar]
- 75. Kotula JW, Kerns SJ, Shaket LA, Siraj L, Collins JJ, Way JC, Silver PA. 2014. Programmable bacteria detect and record an environmental signal in the mammalian gut. Proc Natl Acad Sci U S A 111:4838–4843. doi: 10.1073/pnas.1321321111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Barreto HC, Sousa A, Gordo I. 2020. The landscape of adaptive evolution of a gut commensal bacteria in aging mice. Curr Biol 30:1102–1109. doi: 10.1016/j.cub.2020.01.037 [DOI] [PubMed] [Google Scholar]
- 77. Chen DW, Garud NR. 2022. Rapid evolution and strain turnover in the infant gut microbiome. Genome Res 32:1124–1136. doi: 10.1101/gr.276306.121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Ju F, Beck K, Yin X, Maccagnan A, McArdell CS, Singer HP, Johnson DR, Zhang T, Bürgmann H. 2019. Wastewater treatment plant resistomes are shaped by bacterial composition, genetic exchange, and upregulated expression in the effluent microbiomes. ISME J 13:346–360. doi: 10.1038/s41396-018-0277-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yilmaz B, Mooser C, Keller I, Li H, Zimmermann J, Bosshard L, Fuhrer T, Gomez de Agüero M, Trigo NF, Tschanz-Lischer H, Limenitakis JP, Hardt W-D, McCoy KD, Stecher B, Excoffier L, Sauer U, Ganal-Vonarburg SC, Macpherson AJ. 2021. Long-term evolution and short-term adaptation of microbiota strains and sub-strains in mice. Cell Host Microbe 29:650–663. doi: 10.1016/j.chom.2021.02.001 [DOI] [PubMed] [Google Scholar]
- 80. Matias Rodrigues JF, Schmidt TSB, Tackmann J, von Mering C. 2017. MAPseq: highly efficient k-mer search with confidence estimates, for rRNA sequence analysis. Bioinformatics 33:3808–3810. doi: 10.1093/bioinformatics/btx517 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Johnson JS, Spakowicz DJ, Hong B-Y, Petersen LM, Demkowicz P, Chen L, Leopold SR, Hanson BM, Agresta HO, Gerstein M, Sodergren E, Weinstock GM. 2019. Evaluation of 16S rRNA gene sequencing for species and strain-level microbiome analysis. Nat Commun 10:5029. doi: 10.1038/s41467-019-13036-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Douglas GM, Maffei VJ, Zaneveld JR, Yurgel SN, Brown JR, Taylor CM, Huttenhower C, Langille MGI. 2020. PICRUSt2 for prediction of metagenome functions. Nat Biotechnol 38:685–688. doi: 10.1038/s41587-020-0548-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Goussarov G, Mysara M, Vandamme P, Van Houdt R. 2022. Introduction to the principles and methods underlying the recovery of metagenome-assembled genomes from metagenomic data. Microbiologyopen 11:e1298. doi: 10.1002/mbo3.1298 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Meyer F, Fritz A, Deng Z-L, Koslicki D, Lesker TR, Gurevich A, Robertson G, Alser M, Antipov D, Beghini F, Bertrand D, Brito JJ, Brown CT, Buchmann J, Buluç A, Chen B, Chikhi R, Clausen P, Cristian A, Dabrowski PW, Darling AE, Egan R, Eskin E, Georganas E, Goltsman E, Gray MA, Hansen LH, Hofmeyr S, Huang P, Irber L, Jia H, Jørgensen TS, Kieser SD, Klemetsen T, Kola A, Kolmogorov M, Korobeynikov A, Kwan J, LaPierre N, Lemaitre C, Li C, Limasset A, Malcher-Miranda F, Mangul S, Marcelino VR, Marchet C, Marijon P, Meleshko D, Mende DR, Milanese A, Nagarajan N, Nissen J, Nurk S, Oliker L, Paoli L, Peterlongo P, Piro VC, Porter JS, Rasmussen S, Rees ER, Reinert K, Renard B, Robertsen EM, Rosen GL, Ruscheweyh H-J, Sarwal V, Segata N, Seiler E, Shi L, Sun F, Sunagawa S, Sørensen SJ, Thomas A, Tong C, Trajkovski M, Tremblay J, Uritskiy G, Vicedomini R, Wang Z, Wang Z, Wang Z, Warren A, Willassen NP, Yelick K, You R, Zeller G, Zhao Z, Zhu S, Zhu J, Garrido-Oter R, Gastmeier P, Hacquard S, Häußler S, Khaledi A, Maechler F, Mesny F, Radutoiu S, Schulze-Lefert P, Smit N, Strowig T, Bremges A, Sczyrba A, McHardy AC. 2022. Critical assessment of metagenome interpretation: the second round of challenges. Nat Methods 19:429–440. doi: 10.1038/s41592-022-01431-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ruscheweyh HJ, Milanese A, Paoli L, Karcher N, Clayssen Q, Keller MI, Wirbel J, Bork P, Mende DR, Zeller G, Sunagawa S. 2022. Cultivation-independent genomes greatly expand taxonomic-profiling capabilities of mOTUs across various environments. Microbiome 10:212. doi: 10.1186/s40168-022-01410-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Sunagawa S, Mende DR, Zeller G, Izquierdo-Carrasco F, Berger SA, Kultima JR, Coelho LP, Arumugam M, Tap J, Nielsen HB, Rasmussen S, Brunak S, Pedersen O, Guarner F, de Vos WM, Wang J, Li J, Doré J, Ehrlich SD, Stamatakis A, Bork P. 2013. Metagenomic species profiling using universal phylogenetic marker genes. Nat Methods 10:1196–1199. doi: 10.1038/nmeth.2693 [DOI] [PubMed] [Google Scholar]
- 87. Bickhart DM, Kolmogorov M, Tseng E, Portik DM, Korobeynikov A, Tolstoganov I, Uritskiy G, Liachko I, Sullivan ST, Shin SB, Zorea A, Andreu VP, Panke-Buisse K, Medema MH, Mizrahi I, Pevzner PA, Smith TPL. 2022. Generating lineage-resolved, complete metagenome-assembled genomes from complex microbial communities. Nat Biotechnol 40:711–719. doi: 10.1038/s41587-021-01130-z [DOI] [PubMed] [Google Scholar]
- 88. Tkacz A, Hortala M, Poole PS. 2018. Absolute quantitation of microbiota abundance in environmental samples. Microbiome 6:110. doi: 10.1186/s40168-018-0491-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Vandeputte D, Kathagen G, D’hoe K, Vieira-Silva S, Valles-Colomer M, Sabino J, Wang J, Tito RY, De Commer L, Darzi Y, Vermeire S, Falony G, Raes J. 2017. Quantitative microbiome profiling links gut community variation to microbial load. Nature 551:507–511. doi: 10.1038/nature24460 [DOI] [PubMed] [Google Scholar]
- 90. Özel Duygan BD, Hadadi N, Babu AF, Seyfried M, van der Meer JR. 2020. Rapid detection of microbiota cell type diversity using machine-learned classification of flow cytometry data. Commun Biol 3:379. doi: 10.1038/s42003-020-1106-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Özel Duygan BD, Rey S, Leocata S, Baroux L, Seyfried M, van der Meer JR, Hug LA, Spain J. 2021. Assessing biodegradability of chemical compounds from microbial community growth using flow cytometry. mSystems 6:e01143-20. doi: 10.1128/mSystems.01143-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Röttjers L, Faust K. 2018. From hairballs to hypotheses-biological insights from microbial networks. FEMS Microbiol Rev 42:761–780. doi: 10.1093/femsre/fuy030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Faust K, Lahti L, Gonze D, de Vos WM, Raes J. 2015. Metagenomics meets time series analysis: unraveling microbial community dynamics. Curr Opin Microbiol 25:56–66. doi: 10.1016/j.mib.2015.04.004 [DOI] [PubMed] [Google Scholar]
- 94. Jiang MZ, Zhu HZ, Zhou N, Liu C, Jiang CY, Wang Y, Liu SJ. 2022. Droplet microfluidics-based high-throughput bacterial cultivation for validation of taxon pairs in microbial co-occurrence networks. Sci Rep 12:18145. doi: 10.1038/s41598-022-23000-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Faust K, Raes J. 2012. Microbial interactions: from networks to models. Nat Rev Microbiol 10:538–550. doi: 10.1038/nrmicro2832 [DOI] [PubMed] [Google Scholar]
- 96. Tackmann J, Matias Rodrigues JF, von Mering C. 2019. Rapid inference of direct interactions in large-scale ecological networks from heterogeneous microbial sequencing data. Cell Syst 9:286–296. doi: 10.1016/j.cels.2019.08.002 [DOI] [PubMed] [Google Scholar]
- 97. Berry D, Widder S. 2014. Deciphering microbial interactions and detecting keystone species with co-occurrence networks. Front Microbiol 5:219. doi: 10.3389/fmicb.2014.00219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Stegen JC, Lin X, Fredrickson JK, Konopka AE. 2015. Estimating and mapping ecological processes influencing microbial community assembly. Front Microbiol 6:370. doi: 10.3389/fmicb.2015.00370 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Bucci V, Tzen B, Li N, Simmons M, Tanoue T, Bogart E, Deng L, Yeliseyev V, Delaney ML, Liu Q, Olle B, Stein RR, Honda K, Bry L, Gerber GK. 2016. MDSINE: microbial dynamical systems INference engine for microbiome time-series analyses. Genome Biol 17:121. doi: 10.1186/s13059-016-0980-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Gonze D, Coyte KZ, Lahti L, Faust K. 2018. Microbial communities as dynamical systems. Curr Opin Microbiol 44:41–49. doi: 10.1016/j.mib.2018.07.004 [DOI] [PubMed] [Google Scholar]
- 101. Hoces D, Lan J, Sun W, Geiser T, Stäubli ML, Cappio Barazzone E, Arnoldini M, Challa TD, Klug M, Kellenberger A, Nowok S, Faccin E, Macpherson AJ, Stecher B, Sunagawa S, Zenobi R, Hardt W-D, Wolfrum C, Slack E. 2022. Metabolic reconstitution of germ-free mice by a gnotobiotic microbiota varies over the circadian cycle. PLoS Biol 20:e3001743. doi: 10.1371/journal.pbio.3001743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Peredo EL, Simmons SL. 2017. Leaf-FISH: microscale imaging of bacterial taxa on phyllosphere. Front Microbiol 8:2669. doi: 10.3389/fmicb.2017.02669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Weissbrodt DG, Neu TR, Kuhlicke U, Rappaz Y, Holliger C. 2013. Assessment of bacterial and structural dynamics in aerobic granular biofilms. Front Microbiol 4:175. doi: 10.3389/fmicb.2013.00175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Mark Welch JL, Hasegawa Y, McNulty NP, Gordon JI, Borisy GG. 2017. Spatial organization of a model 15-member human gut microbiota established in gnotobiotic mice. Proc Natl Acad Sci U S A 114:E9105–E9114. doi: 10.1073/pnas.1711596114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Shi H, Shi Q, Grodner B, Lenz JS, Zipfel WR, Brito IL, De Vlaminck I. 2020. Highly multiplexed spatial mapping of microbial communities. Nature 588:676–681. doi: 10.1038/s41586-020-2983-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Cao Z, Zuo W, Wang L, Chen J, Qu Z, Jin F, Dai L. 2023. Spatial profiling of microbial communities by sequential FISH with error-robust encoding. Nat Commun 14:1477. doi: 10.1038/s41467-023-37188-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Dar D, Dar N, Cai L, Newman DK. 2021. Spatial transcriptomics of planktonic and sessile bacterial populations at single-cell resolution. Science 373:eabi4882. doi: 10.1126/science.abi4882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Moor K, Diard M, Sellin ME, Felmy B, Wotzka SY, Toska A, Bakkeren E, Arnoldini M, Bansept F, Co AD, Völler T, Minola A, Fernandez-Rodriguez B, Agatic G, Barbieri S, Piccoli L, Casiraghi C, Corti D, Lanzavecchia A, Regoes RR, Loverdo C, Stocker R, Brumley DR, Hardt W-D, Slack E. 2017. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 544:498–502. doi: 10.1038/nature22058 [DOI] [PubMed] [Google Scholar]
- 109. Müller AJ, Kaiser P, Dittmar KEJ, Weber TC, Haueter S, Endt K, Songhet P, Zellweger C, Kremer M, Fehling H-J, Hardt W-D. 2012. Salmonella gut invasion involves TTSS-2-dependent epithelial traversal, basolateral exit, and uptake by epithelium-sampling lamina propria phagocytes. Cell Host Microbe 11:19–32. doi: 10.1016/j.chom.2011.11.013 [DOI] [PubMed] [Google Scholar]
- 110. Furter M, Sellin ME, Hansson GC, Hardt WD. 2019. Mucus architecture and near-surface swimming affect distinct Salmonella Typhimurium infection patterns along the murine intestinal tract. Cell Rep 27:2665–2678. doi: 10.1016/j.celrep.2019.04.106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Hofer M, Lutolf MP. 2021. Engineering organoids. Nat Rev Mater 6:402–420. doi: 10.1038/s41578-021-00279-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Zhou J, Ning D. 2017. Stochastic community assembly: does it matter in microbial ecology? Microbiol Mol Biol Rev 81:e00002-17. doi: 10.1128/MMBR.00002-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Sprockett D, Fukami T, Relman DA. 2018. Role of priority effects in the early-life assembly of the gut microbiota. Nat Rev Gastroenterol Hepatol 15:197–205. doi: 10.1038/nrgastro.2017.173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Cordovez V, Dini-Andreote F, Carrión VJ, Raaijmakers JM. 2019. Ecology and evolution of plant microbiomes. Annu. Rev. Microbiol 73:69–88. doi: 10.1146/annurev-micro-090817-062524 [DOI] [PubMed] [Google Scholar]
- 115. Hu D, Fuller NR, Caterson ID, Holmes AJ, Reeves PR. 2022. Single-gene long-read sequencing illuminates Escherichia coli strain dynamics in the human intestinal microbiome. Cell Rep 38:110239. doi: 10.1016/j.celrep.2021.110239 [DOI] [PubMed] [Google Scholar]
- 116. Li E, Zhang H, Jiang H, Pieterse CMJ, Jousset A, Bakker P, de Jonge R, Lindow SE, Ausubel FM. 2021. Experimental-evolution-driven identification of Arabidopsis rhizosphere competence genes in Pseudomonas protegens. mBio 12:e0092721. doi: 10.1128/mBio.00927-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Li E, de Jonge R, Liu C, Jiang H, Friman V-P, Pieterse CMJ, Bakker PAHM, Jousset A. 2021. Rapid evolution of bacterial mutualism in the plant rhizosphere. Nat Commun 12:3829. doi: 10.1038/s41467-021-24005-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Saunders AM, Albertsen M, Vollertsen J, Nielsen PH. 2016. The activated sludge ecosystem contains a core community of abundant organisms. ISME J 10:11–20. doi: 10.1038/ismej.2015.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Mallon Cyrus Alexander, Elsas JD van, Salles JF. 2015. Microbial invasions: the process, patterns, and mechanisms. Trends Microbiol 23:719–729. doi: 10.1016/j.tim.2015.07.013 [DOI] [PubMed] [Google Scholar]
- 120. Herzog MK-M, Cazzaniga M, Peters A, Shayya N, Beldi L, Hapfelmeier S, Heimesaat MM, Bereswill S, Frankel G, Gahan CGM, Hardt W-D. 2023. Mouse models for bacterial enteropathogen infections: insights into the role of colonization resistance. Gut Microbes 15:2172667. doi: 10.1080/19490976.2023.2172667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Mallon Cyrus A, Poly F, Le Roux X, Marring I, van Elsas JD, Salles JF. 2015. Resource pulses can alleviate the biodiversity-invasion relationship in soil microbial communities. Ecology 96:915–926. doi: 10.1890/14-1001.1 [DOI] [PubMed] [Google Scholar]
- 122. van Elsas JD, Chiurazzi M, Mallon CA, Elhottova D, Kristufek V, Salles JF. 2012. Microbial diversity determines the invasion of soil by a bacterial pathogen. Proc Natl Acad Sci U S A 109:1159–1164. doi: 10.1073/pnas.1109326109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Litchman E. 2010. Invisible invaders: non-pathogenic invasive microbes in aquatic and terrestrial ecosystems. Ecol Lett 13:1560–1572. doi: 10.1111/j.1461-0248.2010.01544.x [DOI] [PubMed] [Google Scholar]
- 124. Martiny JBH, Bohannan BJM, Brown JH, Colwell RK, Fuhrman JA, Green JL, Horner-Devine MC, Kane M, Krumins JA, Kuske CR, Morin PJ, Naeem S, Øvreås L, Reysenbach A-L, Smith VH, Staley JT. 2006. Microbial biogeography: putting microorganisms on the map. Nat Rev Microbiol 4:102–112. doi: 10.1038/nrmicro1341 [DOI] [PubMed] [Google Scholar]
- 125. Huus KE, Bauer KC, Brown EM, Bozorgmehr T, Woodward SE, Serapio-Palacios A, Boutin RCT, Petersen C, Finlay BB. 2020. Commensal bacteria modulate immunoglobulin A binding in response to host nutrition. Cell Host Microbe 27:909–921. doi: 10.1016/j.chom.2020.03.012 [DOI] [PubMed] [Google Scholar]
- 126. Dapa T, Ramiro RS, Pedro MF, Gordo I, Xavier KB. 2022. Diet leaves a genetic signature in a keystone member of the gut microbiota. Cell Host Microbe 30:183–199. doi: 10.1016/j.chom.2022.01.002 [DOI] [PubMed] [Google Scholar]
- 127. Laganenka L, Lee J-W, Malfertheiner L, Dieterich CL, Fuchs L, Piel J, von Mering C, Sourjik V, Hardt W-D. 2023. Chemotaxis and autoinducer-2 signalling mediate colonization and contribute to co-existence of Escherichia coli strains in the murine gut. Nat Microbiol 8:204–217. doi: 10.1038/s41564-022-01286-7 [DOI] [PubMed] [Google Scholar]
- 128. Keegstra JM, Carrara F, Stocker R. 2022. The ecological roles of bacterial chemotaxis. Nat Rev Microbiol 20:491–504. doi: 10.1038/s41579-022-00709-w [DOI] [PubMed] [Google Scholar]
- 129. Tecon R, Or D. 2017. Biophysical processes supporting the diversity of microbial life in soil. FEMS Microbiol Rev 41:599–623. doi: 10.1093/femsre/fux039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Pion M, Bshary R, Bindschedler S, Filippidou S, Wick LY, Job D, Junier P. 2013. Gains of bacterial flagellar motility in a fungal world. Appl Environ Microbiol 79:6862–6867. doi: 10.1128/AEM.01393-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Kohlmeier S, Smits THM, Ford RM, Keel C, Harms H, Wick LY. 2005. Taking the fungal highway: mobilization of pollutant-degrading bacteria by fungi. Environ Sci Technol 39:4640–4646. doi: 10.1021/es047979z [DOI] [PubMed] [Google Scholar]
- 132. Nemergut DR, Schmidt SK, Fukami T, O’Neill SP, Bilinski TM, Stanish LF, Knelman JE, Darcy JL, Lynch RC, Wickey P, Ferrenberg S. 2013. Patterns and processes of microbial community assembly. Microbiol Mol Biol Rev 77:342–356. doi: 10.1128/MMBR.00051-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Moeller AH, Suzuki TA, Phifer-Rixey M, Nachman MW. 2018. Transmission modes of the mammalian gut microbiota. Science 362:453–457. doi: 10.1126/science.aat7164 [DOI] [PubMed] [Google Scholar]
- 134. Estes AM, Hearn DJ, Snell-Rood EC, Feindler M, Feeser K, Abebe T, Dunning Hotopp JC, Moczek AP. 2013. Brood ball-mediated transmission of microbiome members in the dung beetle, Onthophagus taurus (Coleoptera: Scarabaeidae). PLoS One 8:e79061. doi: 10.1371/journal.pone.0079061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Gudbergsen H, Henriksen M, Wæhrens EE, Overgaard A, Bliddal H, Christensen R, Boesen MP, Knop FKK, Astrup A, Rasmussen MU, Bartholdy C, Daugaard C, Bartels EM, Ellegaard K, Heitmann BL, Kristensen LE. 2019. Effect of liraglutide on body weight and pain in patients with overweight and knee osteoarthritis: protocol for a randomised, double-blind, placebo-controlled, parallel-group, single-centre trial. BMJ Open 9:e024065. doi: 10.1136/bmjopen-2018-024065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Segura Munoz RR, Mantz S, Martínez I, Li F, Schmaltz RJ, Pudlo NA, Urs K, Martens EC, Walter J, Ramer-Tait AE. 2022. Experimental evaluation of ecological principles to understand and modulate the outcome of bacterial strain competition in gut microbiomes. ISME J 16:1681–1682. doi: 10.1038/s41396-022-01227-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Berry D, Mader E, Lee TK, Woebken D, Wang Y, Zhu D, Palatinszky M, Schintlmeister A, Schmid MC, Hanson BT, Shterzer N, Mizrahi I, Rauch I, Decker T, Bocklitz T, Popp J, Gibson CM, Fowler PW, Huang WE, Wagner M. 2015. Tracking heavy water (D2O) incorporation for identifying and sorting active microbial cells. Proc Natl Acad Sci U S A 112:E194–E203. doi: 10.1073/pnas.1420406112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Piccardi P, Vessman B, Mitri S. 2019. Toxicity drives facilitation between 4 bacterial species. Proc Natl Acad Sci U S A 116:15979–15984. doi: 10.1073/pnas.1906172116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Springael D, Peys K, Ryngaert A, Van Roy S, Hooyberghs L, Ravatn R, Heyndrickx M, van der Meer J-R, Vandecasteele C, Mergeay M, Diels L. 2002. Community shifts in a seeded 3-chlorobenzoate degrading membrane biofilm reactor: indications for involvement of in situ horizontal transfer of the clc-element from inoculum to contaminant bacteria. Environ Microbiol 4:70–80. doi: 10.1046/j.1462-2920.2002.00267.x [DOI] [PubMed] [Google Scholar]
- 140. Peters JM, Koo BM, Patino R, Heussler GE, Hearne CC, Qu J, Inclan YF, Hawkins JS, Lu CHS, Silvis MR, Harden MM, Osadnik H, Peters JE, Engel JN, Dutton RJ, Grossman AD, Gross CA, Rosenberg OS. 2019. Enabling genetic analysis of diverse bacteria with Mobile-CRISPRi. Nat Microbiol 4:244–250. doi: 10.1038/s41564-018-0327-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Qu J, Prasad NK, Yu MA, Chen S, Lyden A, Herrera N, Silvis MR, Crawford E, Looney MR, Peters JM, Rosenberg OS. 2019. Modulating pathogenesis with Mobile-CRISPRi. J Bacteriol 201:e00304-19. doi: 10.1128/JB.00304-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. López-Igual R, Bernal-Bayard J, Rodríguez-Patón A, Ghigo J-M, Mazel D. 2019. Engineered toxin-Intein antimicrobials can selectively target and kill antibiotic-resistant bacteria in mixed populations. Nat Biotechnol 37:755–760. doi: 10.1038/s41587-019-0105-3 [DOI] [PubMed] [Google Scholar]
- 143. Pfister SP, Schären OP, Beldi L, Printz A, Notter MD, Mukherjee M, Li H, Limenitakis JP, Werren JP, Tandon D, Cuenca M, Hagemann S, Uster SS, Terrazos MA, Gomez de Agüero M, Schürch CM, Coelho FM, Curtiss R III, Slack E, Balmer ML, Hapfelmeier S. 2020. Uncoupling of invasive bacterial mucosal Immunogenicity from pathogenicity. Nat Commun 11:1978. doi: 10.1038/s41467-020-15891-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Chan CTY, Lee JW, Cameron DE, Bashor CJ, Collins JJ. 2016. 'Deadman' and 'Passcode' microbial kill switches for bacterial containment. Nat Chem Biol 12:82–86. doi: 10.1038/nchembio.1979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Ronchel MC, Ramos JL. 2001. Dual system to reinforce biological containment of recombinant bacteria designed for rhizoremediation. Appl Environ Microbiol 67:2649–2656. doi: 10.1128/AEM.67.6.2649-2656.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Button JE, Autran CA, Reens AL, Cosetta CM, Smriga S, Ericson M, Pierce JV, Cook DN, Lee ML, Sun AK, Alousi AM, Koh AY, Rechtman DJ, Jenq RR, McKenzie GJ. 2022. 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. doi: 10.1016/j.chom.2022.04.001 [DOI] [PubMed] [Google Scholar]
- 147. Scheublin TR, Deusch S, Moreno-Forero SK, Müller JA, van der Meer JR, Leveau JHJ. 2014. Transcriptional profiling of gram-positive Arthrobacter in the phyllosphere: induction of pollutant degradation genes by natural plant phenolic compounds. Environ Microbiol 16:2212–2225. doi: 10.1111/1462-2920.12375 [DOI] [PubMed] [Google Scholar]
- 148. Kim W, Levy SB. 2008. Increased fitness of Pseudomonas fluorescens Pf0-1 leucine auxotrophs in soil. Appl Environ Microbiol 74:3644–3651. doi: 10.1128/AEM.00429-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Seif Y, Choudhary KS, Hefner Y, Anand A, Yang L, Palsson BO. 2020. Metabolic and genetic basis for auxotrophies in gram-negative species. Proc Natl Acad Sci U S A 117:6264–6273. doi: 10.1073/pnas.1910499117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Durack J, Lynch SV. 2019. The gut microbiome: relationships with disease and opportunities for therapy. J Exp Med 216:20–40. doi: 10.1084/jem.20180448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Aggarwala V, Mogno I, Li Z, Yang C, Britton GJ, Chen-Liaw A, Mitcham J, Bongers G, Gevers D, Clemente JC, Colombel J-F, Grinspan A, Faith J. 2021. Precise quantification of bacterial strains after fecal microbiota transplantation delineates long-term engraftment and explains outcomes. Nat Microbiol 6:1309–1318. doi: 10.1038/s41564-021-00966-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Pirnay J-P, Ferry T, Resch G. 2022. Recent progress toward the implementation of phage therapy in western medicine. FEMS Microbiol Rev 46:fuab040. doi: 10.1093/femsre/fuab040 [DOI] [PubMed] [Google Scholar]
- 153. Mu A, McDonald D, Jarmusch AK, Martino C, Brennan C, Bryant M, Humphrey GC, Toronczak J, Schwartz T, Nguyen D, Ackermann G, D’Onofrio A, Strathdee SA, Schooley RT, Dorrestein PC, Knight R, Aslam S. 2021. Assessment of the microbiome during bacteriophage therapy in combination with systemic antibiotics to treat a case of staphylococcal device infection. Microbiome 9:92. doi: 10.1186/s40168-021-01026-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Save J, Que YA, Entenza J, Resch G. 2022. Subtherapeutic doses of vancomycin synergize with bacteriophages for treatment of experimental methicillin-resistant Staphylococcus aureus infective endocarditis. Viruses 14:1792. doi: 10.3390/v14081792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Baaziz H, Baker ZR, Franklin HC, Hsu BB. 2022. Rehabilitation of a misbehaving microbiome: phages for the remodeling of bacterial composition and function. iScience 25:104146. doi: 10.1016/j.isci.2022.104146 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Federici S, Nobs SP, Elinav E. 2021. Phages and their potential to modulate the microbiome and immunity. Cell Mol Immunol 18:889–904. doi: 10.1038/s41423-020-00532-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Eichenseher F, Herpers BL, Badoux P, Leyva-Castillo JM, Geha RS, van der M, McKellar J, Janssen F, de B, Selvakumar L, Rohrig C, Frieling J, Offerhaus M, Loessner MJ, Schmelcher M. 2022. Linker-improved chimeric endolysin selectively kills Staphylococcus aureus in vitro, on reconstituted human epidermis, and in a murine model of skin infection. Antimicrob Agents Chemother 66:e0227321. doi: 10.1128/aac.02273-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Murray E, Draper LA, Ross RP, Hill C. 2021. The advantages and challenges of using endolysins in a clinical setting. Viruses 13:680. doi: 10.3390/v13040680 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Heiman CM, Maurhofer M, Calderon S, Dupasquier M, Marquis J, Keel C, Vacheron J. 2022. Pivotal role of O-antigenic polysaccharide display in the sensitivity against phage tail-like particles in environmental Pseudomonas kin competition. ISME J 16:1683–1693. doi: 10.1038/s41396-022-01217-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Palmer JD, Piattelli E, McCormick BA, Silby MW, Brigham CJ, Bucci V. 2018. Engineered probiotic for the inhibition of Salmonella via tetrathionate-induced production of microcin H47. ACS Infect Dis 4:39–45. doi: 10.1021/acsinfecdis.7b00114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Mortzfeld BM, Palmer JD, Bhattarai SK, Dupre HL, Mercado-Lubio R, Silby MW, Bang C, McCormick BA, Bucci V. 2022. Microcin MccI47 selectively inhibits enteric bacteria and reduces carbapenem-resistant Klebsiella pneumoniae colonization in vivo when administered via an engineered live biotherapeutic. Gut Microbes 14:2127633. doi: 10.1080/19490976.2022.2127633 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Heilbronner S, Krismer B, Brötz-Oesterhelt H, Peschel A. 2021. The microbiome-shaping roles of bacteriocins. Nat Rev Microbiol 19:726–739. doi: 10.1038/s41579-021-00569-w [DOI] [PubMed] [Google Scholar]
- 163. Palmer JD, Mortzfeld BM, Piattelli E, Silby MW, McCormick BA, Bucci V. 2020. Microcin H47: a class IIb microcin with potent activity against multidrug resistant Enterobacteriaceae. ACS Infect Dis 6:672–679. doi: 10.1021/acsinfecdis.9b00302 [DOI] [PubMed] [Google Scholar]
- 164. Zimmermann M, Patil KR, Typas A, Maier L. 2021. Towards a mechanistic understanding of reciprocal drug-microbiome interactions. Mol Syst Biol 17:e10116. doi: 10.15252/msb.202010116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Maier L, Goemans CV, Wirbel J, Kuhn M, Eberl C, Pruteanu M, Müller P, Garcia-Santamarina S, Cacace E, Zhang B, Gekeler C, Banerjee T, Anderson EE, Milanese A, Löber U, Forslund SK, Patil KR, Zimmermann M, Stecher B, Zeller G, Bork P, Typas A. 2021. Unravelling the collateral damage of antibiotics on gut bacteria. Nature 599:120–124. doi: 10.1038/s41586-021-03986-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Delgado-Baquerizo M, Oliverio AM, Brewer TE, Benavent-González A, Eldridge DJ, Bardgett RD, Maestre FT, Singh BK, Fierer N. 2018. A global atlas of the dominant bacteria found in soil. Science 359:320–325. doi: 10.1126/science.aap9516 [DOI] [PubMed] [Google Scholar]
- 167. Freter R, Brickner H, Fekete J, Vickerman MM, Carey KE. 1983. Survival and implantation of Escherichia coli in the intestinal tract. Infect Immun 39:686–703. doi: 10.1128/iai.39.2.686-703.1983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Kreuzer M, Hardt WD. 2020. How food affects colonization resistance against enteropathogenic bacteria. Annu Rev Microbiol 74:787–813. doi: 10.1146/annurev-micro-020420-013457 [DOI] [PubMed] [Google Scholar]
- 169. Barthel M, Hapfelmeier S, Quintanilla-Martínez L, Kremer M, Rohde M, Hogardt M, Pfeffer K, Rüssmann H, Hardt W-D. 2003. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar Typhimurium colitis model that allows analysis of both pathogen and host. Infect Immun 71:2839–2858. doi: 10.1128/IAI.71.5.2839-2858.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Diard M, Bakkeren E, Lentsch V, Rocker A, Bekele NA, Hoces D, Aslani S, Arnoldini M, Böhi F, Schumann-Moor K, Adamcik J, Piccoli L, Lanzavecchia A, Stadtmueller BM, Donohue N, van der Woude MW, Hockenberry A, Viollier PH, Falquet L, Wüthrich D, Bonfiglio F, Loverdo C, Egli A, Zandomeneghi G, Mezzenga R, Holst O, Meier BH, Hardt W-D, Slack E. 2021. A rationally designed oral vaccine induces immunoglobulin A in the murine gut that directs the evolution of attenuated Salmonella variants. Nat Microbiol 6:830–841. doi: 10.1038/s41564-021-00911-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Akhiani AA, Stensson A, Schön K, Lycke NY. 2005. IgA antibodies impair resistance against Helicobacter pylori infection: studies on immune evasion in IL-10-deficient mice. J Immunol 174:8144–8153. doi: 10.4049/jimmunol.174.12.8144 [DOI] [PubMed] [Google Scholar]
- 172. Donaldson GP, Ladinsky MS, Yu KB, Sanders JG, Yoo BB, Chou WC, Conner ME, Earl AM, Knight R, Bjorkman PJ, Mazmanian SK. 2018. Gut microbiota utilize immunoglobulin A for mucosal colonization. Science 360:795–800. doi: 10.1126/science.aaq0926 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Joglekar P, Ding H, Canales-Herrerias P, Pasricha PJ, Sonnenburg JL, Peterson DA. 2019. Intestinal IgA regulates expression of a fructan polysaccharide utilization locus in colonizing gut commensal Bacteroides thetaiotaomicron. mBio 10:e02324–02319. doi: 10.1128/mBio.02324-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Nakajima A, Vogelzang A, Maruya M, Miyajima M, Murata M, Son A, Kuwahara T, Tsuruyama T, Yamada S, Matsuura M, Nakase H, Peterson DA, Fagarasan S, Suzuki K. 2018. IgA regulates the composition and metabolic function of gut microbiota by promoting symbiosis between bacteria. J Exp Med 215:2019–2034. doi: 10.1084/jem.20180427 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Peterson DA, Planer JD, Guruge JL, Xue L, Downey-Virgin W, Goodman AL, Seedorf H, Gordon JI. 2015. Characterizing the interactions between a naturally primed immunoglobulin A and its conserved Bacteroides thetaiotaomicron species-specific epitope in gnotobiotic mice. J Biol Chem 290:12630–12649. doi: 10.1074/jbc.M114.633800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. O’Brien EJ, Monk JM, Palsson BO. 2015. Using genome-scale models to predict biological capabilities. Cell 161:971–987. doi: 10.1016/j.cell.2015.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Salvy P, Fengos G, Ataman M, Pathier T, Soh KC, Hatzimanikatis V. 2019. pyTFA and matTFA: a python package and a matlab toolbox for thermodynamics-based flux analysis. Bioinformatics 35:167–169. doi: 10.1093/bioinformatics/bty499 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Oftadeh O, Salvy P, Masid M, Curvat M, Miskovic L, Hatzimanikatis V. 2021. A genome-scale metabolic model of Saccharomyces cerevisiae that integrates expression constraints and reaction thermodynamics. Nat Commun 12:4790. doi: 10.1038/s41467-021-25158-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Salvy P, Hatzimanikatis V. 2021. Emergence of diauxie as an optimal growth strategy under resource allocation constraints in cellular metabolism. Proc Natl Acad Sci U S A 118:e2013836118. doi: 10.1073/pnas.2013836118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. van Berlo RJP, de Ridder D, Daran J-M, Daran-Lapujade PAS, Teusink B, Reinders MJT. 2011. Predicting metabolic fluxes using gene expression differences as constraints. IEEE/ACM Trans Comput Biol Bioinform 8:206–216. doi: 10.1109/TCBB.2009.55 [DOI] [PubMed] [Google Scholar]
- 181. Covert MW, Schilling CH, Palsson B. 2001. Regulation of gene expression in flux balance models of metabolism. J Theor Biol 213:73–88. doi: 10.1006/jtbi.2001.2405 [DOI] [PubMed] [Google Scholar]
- 182. Miskovic L, Hatzimanikatis V. 2010. Production of biofuels and biochemicals: in need of an ORACLE. Trends Biotechnol 28:391–397. doi: 10.1016/j.tibtech.2010.05.003 [DOI] [PubMed] [Google Scholar]
- 183. Ataman M, Hernandez Gardiol DF, Fengos G, Hatzimanikatis V. 2017. redGEM: systematic reduction and analysis of genome-scale metabolic reconstructions for development of consistent core metabolic models. PLoS Comput Biol 13:e1005444. doi: 10.1371/journal.pcbi.1005444 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Ataman M, Hatzimanikatis V. 2017. lumpGEM: systematic generation of subnetworks and elementally balanced lumped reactions for the biosynthesis of target metabolites. PLoS Comput Biol 13:e1005513. doi: 10.1371/journal.pcbi.1005513 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Hafner J, Hatzimanikatis V. 2021. Finding metabolic pathways in large networks through atom-conserving substrate-product pairs. Bioinformatics 37:3560–3568. doi: 10.1093/bioinformatics/btab368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. MohammadiPeyhani H, Hafner J, Sveshnikova A, Viterbo V, Hatzimanikatis V. 2022. Expanding biochemical knowledge and illuminating metabolic dark matter with ATLASx. Nat Commun 13:1560. doi: 10.1038/s41467-022-29238-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Hadadi N, MohammadiPeyhani H, Miskovic L, Seijo M, Hatzimanikatis V. 2019. Enzyme annotation for orphan and novel reactions using knowledge of substrate reactive sites. Proc Natl Acad Sci U S A 116:7298–7307. doi: 10.1073/pnas.1818877116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Glöckler M, Dräger A, Mostolizadeh R. 2022. NCMW: a python package to analyze metabolic interactions in the nasal microbiome. Front Bioinform 2:827024. doi: 10.3389/fbinf.2022.827024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Klitgord N, Segrè D. 2010. Environments that induce synthetic microbial ecosystems. PLoS Comput Biol 6:e1001002. doi: 10.1371/journal.pcbi.1001002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zelezniak A, Andrejev S, Ponomarova O, Mende DR, Bork P, Patil KR. 2015. Metabolic dependencies drive species co-occurrence in diverse microbial communities. Proc Natl Acad Sci U S A 112:6449–6454. doi: 10.1073/pnas.1421834112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Dubinkina V, Fridman Y, Pandey PP, Maslov S. 2019. Multistability and regime shifts in microbial communities explained by competition for essential nutrients. Elife 8:e49720. doi: 10.7554/eLife.49720 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Wang Y, Ye J, Ju F, Liu L, Boyd JA, Deng Y, Parks DH, Jiang X, Yin X, Woodcroft BJ, Tyson GW, Hugenholtz P, Polz MF, Zhang T. 2021. Successional dynamics and alternative stable states in a saline activated sludge microbial community over 9 years. Microbiome 9:199. doi: 10.1186/s40168-021-01151-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Khandelwal RA, Olivier BG, Röling WFM, Teusink B, Bruggeman FJ. 2013. Community flux balance analysis for microbial consortia at balanced growth. PLoS One 8:e64567. doi: 10.1371/journal.pone.0064567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Zomorrodi AR, Maranas CD. 2012. OptCom: a multi-level optimization framework for the metabolic modeling and analysis of microbial communities. PLoS Comput Biol 8:e1002363. doi: 10.1371/journal.pcbi.1002363 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Stolyar S, Van Dien S, Hillesland KL, Pinel N, Lie TJ, Leigh JA, Stahl DA. 2007. Metabolic modeling of a mutualistic microbial community. Mol Syst Biol 3:92. doi: 10.1038/msb4100131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Chan SHJ, Simons MN, Maranas CD. 2017. SteadyCom: predicting microbial abundances while ensuring community stability. PLoS Comput Biol 13:e1005539. doi: 10.1371/journal.pcbi.1005539 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Lloyd CJ, King ZA, Sandberg TE, Hefner Y, Olson CA, Phaneuf PV, O’Brien EJ, Sanders JG, Salido RA, Sanders K, Brennan C, Humphrey G, Knight R, Feist AM, Hatzimanikatis V. 2019. The genetic basis for adaptation of model-designed syntrophic co-cultures. PLoS Comput Biol 15:e1006213. doi: 10.1371/journal.pcbi.1006213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Xu X, Zarecki R, Medina S, Ofaim S, Liu X, Chen C, Hu S, Brom D, Gat D, Porob S, Eizenberg H, Ronen Z, Jiang J, Freilich S. 2019. Modeling microbial communities from atrazine contaminated soils promotes the development of biostimulation solutions. ISME J 13:494–508. doi: 10.1038/s41396-018-0288-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Tobalina L, Bargiela R, Pey J, Herbst F-A, Lores I, Rojo D, Barbas C, Peláez AI, Sánchez J, von Bergen M, Seifert J, Ferrer M, Planes FJ. 2015. Context-specific metabolic network reconstruction of a naphthalene-degrading bacterial community guided by metaproteomic data. Bioinformatics 31:1771–1779. doi: 10.1093/bioinformatics/btv036 [DOI] [PubMed] [Google Scholar]
- 200. Gorter FA, Manhart M, Ackermann M. 2020. Understanding the evolution of interspecies interactions in microbial communities. Philos Trans R Soc Lond B Biol Sci 375:20190256. doi: 10.1098/rstb.2019.0256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. van den Berg NI, Machado D, Santos S, Rocha I, Chacón J, Harcombe W, Mitri S, Patil KR. 2022. Ecological modelling approaches for predicting emergent properties in microbial communities. Nat Ecol Evol 6:855–865. doi: 10.1038/s41559-022-01746-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Zomorrodi AR, Islam MM, Maranas CD. 2014. d-OptCom: dynamic multi-level and multi-objective metabolic modeling of microbial communities. ACS Synth Biol 3:247–257. doi: 10.1021/sb4001307 [DOI] [PubMed] [Google Scholar]
- 203. Zhuang K, Izallalen M, Mouser P, Richter H, Risso C, Mahadevan R, Lovley DR. 2011. Genome-scale dynamic modeling of the competition between rhodoferax and geobacter in anoxic subsurface environments. ISME J 5:305–316. doi: 10.1038/ismej.2010.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Chakrabarti A, Miskovic L, Soh KC, Hatzimanikatis V. 2013. Towards kinetic modeling of genome-scale metabolic networks without sacrificing stoichiometric, thermodynamic and physiological constraints. Biotechnol J 8:1043–1057. doi: 10.1002/biot.201300091 [DOI] [PubMed] [Google Scholar]
- 205. Schäfer M, Pacheco AR, Künzler R, Bortfeld-Miller M, Field CM, Vayena E, Hatzimanikatis V, Vorholt JA. 2023. Metabolic interaction models recapitulate leaf microbiota ecology. Science 381:eadf5121. doi: 10.1126/science.adf5121 [DOI] [PubMed] [Google Scholar]
- 206. Angeles-Martinez L, Hatzimanikatis V. 2021. The influence of the crowding assumptions in biofilm simulations. PLoS Comput Biol 17:e1009158. doi: 10.1371/journal.pcbi.1009158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Kreft J-U, Booth G, Wimpenny JWT. 1998. BacSim, a simulator for individual-based modelling of bacterial colony growth. Microbiology (Reading) 144 ( Pt 12):3275–3287. doi: 10.1099/00221287-144-12-3275 [DOI] [PubMed] [Google Scholar]
- 208. Angeles-Martinez L, Hatzimanikatis V. 2021. Spatio-temporal modeling of the crowding conditions and metabolic variability in microbial communities. PLoS Comput Biol 17:e1009140. doi: 10.1371/journal.pcbi.1009140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Li B, Taniguchi D, Gedara JP, Gogulancea V, Gonzalez-Cabaleiro R, Chen J, McGough AS, Ofiteru ID, Curtis TP, Zuliani P. 2019. NUFEB: a massively parallel simulator for individual-based modelling of microbial communities. PLoS Comput Biol 15:e1007125. doi: 10.1371/journal.pcbi.1007125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Lardon LA, Merkey BV, Martins S, Dötsch A, Picioreanu C, Kreft J-U, Smets BF. 2011. iDynoMiCS: next-generation individual-based modelling of biofilms. Environ Microbiol 13:2416–2434. doi: 10.1111/j.1462-2920.2011.02414.x [DOI] [PubMed] [Google Scholar]
- 211. Phalak P, Chen J, Carlson RP, Henson MA. 2016. Metabolic modeling of a chronic wound biofilm consortium predicts spatial partitioning of bacterial species. BMC Syst Biol 10:90. doi: 10.1186/s12918-016-0334-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Smith WPJ, Davit Y, Osborne JM, Kim W, Foster KR, Pitt-Francis JM. 2017. Cell morphology drives spatial patterning in microbial communities. Proc Natl Acad Sci U S A 114:E280–E286. doi: 10.1073/pnas.1613007114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Dukovski I, Bajić D, Chacón JM, Quintin M, Vila JCC, Sulheim S, Pacheco AR, Bernstein DB, Riehl WJ, Korolev KS, Sanchez A, Harcombe WR, Segrè D. 2021. A metabolic modeling platform for the computation of microbial ecosystems in time and space (COMETS). Nat Protoc 16:5030–5082. doi: 10.1038/s41596-021-00593-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Borer B, Or D. 2021. Spatiotemporal metabolic modeling of bacterial life in complex habitats. Curr Opin Biotechnol 67:65–71. doi: 10.1016/j.copbio.2021.01.004 [DOI] [PubMed] [Google Scholar]
- 215. Jayathilake PG, Gupta P, Li B, Madsen C, Oyebamiji O, González-Cabaleiro R, Rushton S, Bridgens B, Swailes D, Allen B, McGough AS, Zuliani P, Ofiteru ID, Wilkinson D, Chen J, Curtis T. 2017. A mechanistic Individual-based model of microbial communities. PLoS One 12:e0181965. doi: 10.1371/journal.pone.0181965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Nagarajan K, Ni C, Lu T. 2022. Agent-based modeling of microbial communities. ACS Synth Biol 11:3564–3574. doi: 10.1021/acssynbio.2c00411 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Bakker PAHM, Pieterse CMJ, de Jonge R, Berendsen RL. 2018. The soil-borne legacy. Cell 172:1178–1180. doi: 10.1016/j.cell.2018.02.024 [DOI] [PubMed] [Google Scholar]
- 218. Pronk LJU, Bakker PAHM, Keel C, Maurhofer M, Flury P. 2022. The secret life of plant-beneficial rhizosphere bacteria: Insects as alternative hosts. Environ Microbiol 24:3273–3289. doi: 10.1111/1462-2920.15968 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Haskett TL, Tkacz A, Poole PS. 2021. Engineering rhizobacteria for sustainable agriculture. ISME J 15:949–964. doi: 10.1038/s41396-020-00835-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Zhalnina K, Dias R, de Quadros PD, Davis-Richardson A, Camargo FAO, Clark IM, McGrath SP, Hirsch PR, Triplett EW. 2015. Soil pH determines microbial diversity and composition in the park grass experiment. Microb Ecol 69:395–406. doi: 10.1007/s00248-014-0530-2 [DOI] [PubMed] [Google Scholar]
- 221. Bhattacharyya A, Pablo CHD, Mavrodi OV, Weller DM, Thomashow LS, Mavrodi DV. 2021. Rhizosphere plant-microbe interactions under water stress. Adv Appl Microbiol 115:65–113. doi: 10.1016/bs.aambs.2021.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Trivedi P, Leach JE, Tringe SG, Sa T, Singh BK. 2020. Plant-microbiome interactions: from community assembly to plant health. Nat Rev Microbiol 18:607–621. doi: 10.1038/s41579-020-0412-1 [DOI] [PubMed] [Google Scholar]
- 223. Saad MM, Eida AA, Hirt H. 2020. Tailoring plant-associated microbial inoculants in agriculture: a roadmap for successful application. J Exp Bot 71:3878–3901. doi: 10.1093/jxb/eraa111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Fitzpatrick CR, Salas-González I, Conway JM, Finkel OM, Gilbert S, Russ D, Teixeira PJPL, Dangl JL. 2020. The plant microbiome: from ecology to reductionism and beyond. Annu Rev Microbiol 74:81–100. doi: 10.1146/annurev-micro-022620-014327 [DOI] [PubMed] [Google Scholar]
- 225. Bulgarelli D, Schlaeppi K, Spaepen S, Ver Loren van Themaat E, Schulze-Lefert P. 2013. Structure and functions of the bacterial microbiota of plants. Annu Rev Plant Biol 64:807–838. doi: 10.1146/annurev-arplant-050312-120106 [DOI] [PubMed] [Google Scholar]
- 226. Yu P, He X, Baer M, Beirinckx S, Tian T, Moya YAT, Zhang X, Deichmann M, Frey FP, Bresgen V, Li C, Razavi BS, Schaaf G, von Wirén N, Su Z, Bucher M, Tsuda K, Goormachtig S, Chen X, Hochholdinger F. 2021. Plant flavones enrich rhizosphere Oxalobacteraceae to improve maize performance under nitrogen deprivation. Nat Plants 7:481–499. doi: 10.1038/s41477-021-00897-y [DOI] [PubMed] [Google Scholar]
- 227. Cotton TEA, Pétriacq P, Cameron DD, Meselmani MA, Schwarzenbacher R, Rolfe SA, Ton J. 2019. Metabolic regulation of the maize rhizobiome by benzoxazinoids. ISME J 13:1647–1658. doi: 10.1038/s41396-019-0375-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Vismans G, van Bentum S, Spooren J, Song Y, Goossens P, Valls J, Snoek BL, Thiombiano B, Schilder M, Dong L, Bouwmeester HJ, Pétriacq P, Pieterse CMJ, Bakker P, Berendsen RL. 2022. Coumarin biosynthesis genes are required after foliar pathogen infection for the creation of a microbial soil-borne legacy that primes plants for SA-dependent defenses. Sci Rep 12:22473. doi: 10.1038/s41598-022-26551-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Stassen MJJ, Hsu SH, Pieterse CMJ, Stringlis IA. 2021. Coumarin communication along the microbiome-root-shoot axis. Trends Plant Sci 26:169–183. doi: 10.1016/j.tplants.2020.09.008 [DOI] [PubMed] [Google Scholar]
- 230. McRose DL, Newman DK. 2021. Redox-active antibiotics enhance phosphorus bioavailability. Science 371:1033–1037. doi: 10.1126/science.abd1515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. LeTourneau MK, Marshall MJ, Cliff JB, Bonsall RF, Dohnalkova AC, Mavrodi DV, Devi SI, Mavrodi OV, Harsh JB, Weller DM, Thomashow LS. 2018. Phenazine-1-carboxylic acid and soil moisture influence biofilm development and turnover of rhizobacterial biomass on wheat root surfaces. Environ Microbiol 20:2178–2194. doi: 10.1111/1462-2920.14244 [DOI] [PubMed] [Google Scholar]
- 232. Yu K, Liu Y, Tichelaar R, Savant N, Lagendijk E, van Kuijk SJL, Stringlis IA, van Dijken AJH, Pieterse CMJ, Bakker PAHM, Haney CH, Berendsen RL. 2019. Rhizosphere-associated Pseudomonas suppress local root immune responses by gluconic acid-mediated lowering of environmental pH. Curr Biol 29:3913–3920. doi: 10.1016/j.cub.2019.09.015 [DOI] [PubMed] [Google Scholar]
- 233. Dar D, Thomashow LS, Weller DM, Newman DK. 2020. Global landscape of phenazine biosynthesis and biodegradation reveals species-specific colonization patterns in agricultural soils and crop microbiomes. Elife 9:e59726. doi: 10.7554/eLife.59726 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Venturi V, Keel C. 2016. Signaling in the rhizosphere. Trends Plant Sci 21:187–198. doi: 10.1016/j.tplants.2016.01.005 [DOI] [PubMed] [Google Scholar]
- 235. Etalo DW, Jeon JS, Raaijmakers JM. 2018. Modulation of plant chemistry by beneficial root microbiota. Nat Prod Rep 35:398–409. doi: 10.1039/c7np00057j [DOI] [PubMed] [Google Scholar]
- 236. Ravelo-Ortega G, Raya-González J, López-Bucio J. 2023. Compounds from rhizosphere microbes that promote plant growth. Curr Opin Plant Biol 73:102336. doi: 10.1016/j.pbi.2023.102336 [DOI] [PubMed] [Google Scholar]
- 237. Thomashow LS, Kwak YS, Weller DM. 2019. Root-associated microbes in sustainable agriculture: models, metabolites and mechanisms. Pest Manag Sci 75:2360–2367. doi: 10.1002/ps.5406 [DOI] [PubMed] [Google Scholar]
- 238. Boak EN, Kirolos S, Pan H, Pierson LS III, Pierson EA. 2022. The type VI secretion systems in plant-beneficial bacteria modulate prokaryotic and eukaryotic interactions in the rhizosphere. Front. Microbiol 13:843092. doi: 10.3389/fmicb.2022.843092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Ghequire MGK, De Mot R. 2018. Turning over a new leaf: bacteriocins going green. Trends Microbiol 26:1–2. doi: 10.1016/j.tim.2017.11.001 [DOI] [PubMed] [Google Scholar]
- 240. Dorosky RJ, Pierson LS III, Pierson EA, Vieille C. 2018. Pseudomonas chlororaphis produces multiple R-tailocin particles that broaden the killing spectrum and contribute to persistence in rhizosphere communities. Appl Environ Microbiol 84. doi: 10.1128/AEM.01230-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Vacheron J, Heiman CM, Keel C. 2021. Live cell dynamics of production, explosive release and killing activity of phage tail-like weapons for Pseudomonas kin exclusion. Commun Biol 4:87. doi: 10.1038/s42003-020-01581-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Keel C, Ucurum Z, Michaux P, Adrian M, Haas D. 2002. Deleterious impact of a virulent bacteriophage on survival and biocontrol activity of Pseudomonas fluorescens strain CHAO in natural soil. Mol Plant Microbe Interact 15:567–576. doi: 10.1094/MPMI.2002.15.6.567 [DOI] [PubMed] [Google Scholar]
- 243. Pratama AA, Terpstra J, de Oliveria ALM, Salles JF. 2020. The role of rhizosphere bacteriophages in plant health. Trends Microbiol 28:709–718. doi: 10.1016/j.tim.2020.04.005 [DOI] [PubMed] [Google Scholar]
- 244. Gao Z, Karlsson I, Geisen S, Kowalchuk G, Jousset A. 2019. Protists: puppet masters of the rhizosphere microbiome. Trends in Plant Science 24:165–176. doi: 10.1016/j.tplants.2018.10.011 [DOI] [PubMed] [Google Scholar]
- 245. Renoud S, Vacheron J, Abrouk D, Prigent-Combaret C, Legendre L, Muller D, Moënne-Loccoz Y. 2021. Field site-specific effects of an Azospirillum seed inoculant on key microbial functional groups in the rhizosphere. Front Microbiol 12:760512. doi: 10.3389/fmicb.2021.760512 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Renoud S, Abrouk D, Prigent-Combaret C, Wisniewski-Dyé F, Legendre L, Moënne-Loccoz Y, Muller D. 2022. Effect of inoculation level on the impact of the PGPR Azospirillum lipoferum CRT1 on selected microbial functional groups in the rhizosphere of field maize. Microorganisms 10:325. doi: 10.3390/microorganisms10020325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Morales Moreira ZP, Chen MY, Yanez Ortuno DL, Haney CH. 2023. Engineering plant microbiomes by integrating eco-evolutionary principles into current strategies. Curr Opin Plant Biol 71:102316. doi: 10.1016/j.pbi.2022.102316 [DOI] [PubMed] [Google Scholar]
- 248. Carrión VJ, Perez-Jaramillo J, Cordovez V, Tracanna V, de Hollander M, Ruiz-Buck D, Mendes LW, van Ijcken WFJ, Gomez-Exposito R, Elsayed SS, Mohanraju P, Arifah A, van der Oost J, Paulson JN, Mendes R, van Wezel GP, Medema MH, Raaijmakers JM. 2019. Pathogen-induced activation of disease-suppressive functions in the endophytic root microbiome. Science 366:606–612. doi: 10.1126/science.aaw9285 [DOI] [PubMed] [Google Scholar]
- 249. Raaijmakers JM, Mazzola M. 2016. ECOLOGY. Soil immune responses. Science 352:1392–1393. doi: 10.1126/science.aaf3252 [DOI] [PubMed] [Google Scholar]
- 250. Holtappels D, Fortuna K, Lavigne R, Wagemans J. 2021. The future of phage biocontrol in integrated plant protection for sustainable crop production. Curr Opin Biotechnol 68:60–71. doi: 10.1016/j.copbio.2020.08.016 [DOI] [PubMed] [Google Scholar]
- 251. Baltrus DA, Clark M, Hockett KL, Mollico M, Smith C, Weaver S. 2022. Prophylactic application of tailocins prevents infection by Pseudomonas syringae. Phytopathology 112:561–566. doi: 10.1094/PHYTO-06-21-0269-R [DOI] [PubMed] [Google Scholar]
- 252. Rooney WM, Chai R, Milner JJ, Walker D. 2020. Bacteriocins targeting gram-negative phytopathogenic bacteria: plantibiotics of the future. Front Microbiol 11:575981. doi: 10.3389/fmicb.2020.575981 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Príncipe A, Fernandez M, Torasso M, Godino A, Fischer S. 2018. Effectiveness of tailocins produced by Pseudomonas fluorescens SF4c in controlling the bacterial-spot disease in tomatoes caused by Xanthomonas vesicatoria. Microbiol Res 212–213:94–102. doi: 10.1016/j.micres.2018.05.010 [DOI] [PubMed] [Google Scholar]
- 254. Oyserman BO, Flores SS, Griffioen T, Pan X, van der Wijk E, Pronk L, Lokhorst W, Nurfikari A, Paulson JN, Movassagh M, Stopnisek N, Kupczok A, Cordovez V, Carrión VJ, Ligterink W, Snoek BL, Medema MH, Raaijmakers JM. 2022. Disentangling the genetic basis of rhizosphere microbiome assembly in tomato. Nat Commun 13:3228. doi: 10.1038/s41467-022-30849-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Song C, Jin K, Raaijmakers JM. 2021. Designing a home for beneficial plant microbiomes. Curr Opin Plant Biol 62:102025. doi: 10.1016/j.pbi.2021.102025 [DOI] [PubMed] [Google Scholar]
- 256. El Fantroussi S, Agathos SN. 2005. Is bioaugmentation a feasible strategy for pollutant removal and site remediation? Curr Opin Microbiol 8:268–275. doi: 10.1016/j.mib.2005.04.011 [DOI] [PubMed] [Google Scholar]
- 257. van Veen JA, van Overbeek LS, van Elsas JD. 1997. Fate and activity of microorganisms introduced into soil. Microbiol Mol Biol Rev 61:121–135. doi: 10.1128/mmbr.61.2.121-135.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Halden RU, Halden BG, Dwyer DF. 1999. Removal of dibenzofuran, dibenzo-p-dioxin, and 2-chlorodibenzo-p-dioxin from soils inoculated with Sphingomonas sp. strain RW1. Appl Environ Microbiol 65:2246–2249. doi: 10.1128/AEM.65.5.2246-2249.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Atashgahi S, Sánchez-Andrea I, Heipieper HJ, van der Meer JR, Stams AJM, Smidt H. 2018. Prospects for harnessing biocide resistance for bioremediation and detoxification. Science 360:743–746. doi: 10.1126/science.aar3778 [DOI] [PubMed] [Google Scholar]
- 260. Dwyer DF, Rojo F, Timmis KN. 1988. Fate and behaviour in an activated sludge microcosm of a genetically-engineered micro-organism designed to degrade substituted aromatic compounds, p 77–88. In Sussmann M, Collins CH, Skinner FA, Stewart-Tull DE (ed), The release of genetically-engineered micro-organisms. Academic Press, Inc, London. [Google Scholar]
- 261. Pipke R, Wagner-Döbler I, Timmis KN, Dwyer DF. 1992. Survival and function of a genetically engineered pseudomonad in aquatic sediment microcosms. Appl Environ Microbiol 58:1259–1265. doi: 10.1128/aem.58.4.1259-1265.1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. de Lorenzo V, Pieper D, Ramos JL. 2013. From the test tube to the environment - and back. Environ Microbiol 15:6–11. doi: 10.1111/j.1462-2920.2012.02896.x [DOI] [PubMed] [Google Scholar]
- 263. Wagner-Döbler I, Pipke R, Timmis KN, Dwyer DF. 1992. Evaluation of aquatic sediment microcosms and their use in assessing possible effects of introduced microorganisms on ecosystem parameters. Appl Environ Microbiol 58:1249–1258. doi: 10.1128/aem.58.4.1249-1258.1992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. van der Meer JR, Roelofsen W, Schraa G, Zehnder AJB. 1987. Degradation of low concentrations of dichlorobenzenes and 1,2,4-trichlorobenzene by Pseudomonas sp. strain P51 in nonsterile soil columns. FEMS Microbiol. Ecol 45:333–341. doi: 10.1111/j.1574-6968.1987.tb02410.x [DOI] [Google Scholar]
- 265. Tchelet R, Meckenstock R, Steinle P, van der Meer JR. 1999. Population dynamics of an introduced bacterium degrading chlorinated benzenes in a soil column and in sewage sludge. Biodegradation 10:113–125. doi: 10.1023/a:1008368006917 [DOI] [PubMed] [Google Scholar]
- 266. Watanabe K, Teramoto M, Harayama S. 2002. Stable augmentation of activated sludge with foreign catabolic genes harboured by an indigenous dominant bacterium. Environ Microbiol 4:577–583. doi: 10.1046/j.1462-2920.2002.00342.x [DOI] [PubMed] [Google Scholar]
- 267. Roane TM, Josephson KL, Pepper IL. 2001. Dual-bioaugmentation strategy to enhance remediation of cocontaminated soil. Appl Environ Microbiol 67:3208–3215. doi: 10.1128/AEM.67.7.3208-3215.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Bosma TNP, Middeldorp PJM, Schraa G, Zehnder AJB. 1997. Mass transfer limitation of biotransformation: quantifying bioavailability. Environ Sci Technol 31:248–252. doi: 10.1021/es960383u [DOI] [Google Scholar]
- 269. Chai B, Tsoi T, Sallach JB, Liu C, Landgraf J, Bezdek M, Zylstra G, Li H, Johnston CT, Teppen BJ, Cole JR, Boyd SA, Tiedje JM. 2020. Bioavailability of clay-adsorbed dioxin to Sphingomonas wittichii RW1 and its associated genome-wide shifts in gene expression. Sci Total Environ 712:135525. doi: 10.1016/j.scitotenv.2019.135525 [DOI] [PubMed] [Google Scholar]
- 270. Coppotelli BM, Ibarrolaza A, Dias RL, Del Panno MT, Berthe-Corti L, Morelli IS. 2010. Study of the degradation activity and the strategies to promote the bioavailability of phenanthrene by Sphingomonas paucimobilis strain 20006FA. Microb Ecol 59:266–276. doi: 10.1007/s00248-009-9563-3 [DOI] [PubMed] [Google Scholar]
- 271. Morales M, Sentchilo V, Bertelli C, Komljenovic A, Kryuchkova-Mostacci N, Bourdilloud A, Linke B, Goesmann A, Harshman K, Segers F, Delapierre F, Fiorucci D, Seppey M, Trofimenco E, Berra P, El Taher A, Loiseau C, Roggero D, Sulfiotti M, Etienne A, Ruiz Buendia G, Pillard L, Escoriza A, Moritz R, Schneider C, Alfonso E, Ben Jeddou F, Selmoni O, Resch G, Greub G, Emery O, Dubey M, Pillonel T, Robinson-Rechavi M, van der Meer JR. 2016. The genome of the toluene-degrading Pseudomonas veronii strain 1YdBTEX2 and its differential gene expression in contaminated sand. PLoS One 11:e0165850. doi: 10.1371/journal.pone.0165850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Lima-Morales D, Jáuregui R, Camarinha-Silva A, Geffers R, Pieper DH, Vilchez-Vargas R. 2016. Linking microbial community and catabolic gene structures during the adaptation of three contaminated soils under continuous long-term pollutant stress. Appl Environ Microbiol 82:2227–2237. doi: 10.1128/AEM.03482-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Moreno-Forero SK, Rojas E, Beggah S, van der Meer JR. 2016. Comparison of differential gene expression to water stress among bacteria with relevant pollutant-degradation properties. Environ Microbiol Rep 8:91–102. doi: 10.1111/1758-2229.12356 [DOI] [PubMed] [Google Scholar]
- 274. Fida TT, Moreno-Forero SK, Heipieper HJ, Springael D. 2013. Physiology and transcriptome of the polycyclic aromatic hydrocarbon-degrading Sphingomonas sp. LH128 after long-term starvation. Microbiology (Reading) 159:1807–1817. doi: 10.1099/mic.0.065870-0 [DOI] [PubMed] [Google Scholar]
- 275. Svenningsen NB, Pérez-Pantoja D, Nikel PI, Nicolaisen MH, de Lorenzo V, Nybroe O. 2015. Pseudomonas putida mt-2 tolerates reactive oxygen species generated during matric stress by inducing a major oxidative defense response. BMC Microbiol 15:202. doi: 10.1186/s12866-015-0542-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Gülez G, Dechesne A, Workman CT, Smets BF. 2012. Transcriptome dynamics of Pseudomonas putida KT2440 under water stress. Appl Environ Microbiol 78:676–683. doi: 10.1128/AEM.06150-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Moreno-Forero SK, van der Meer JR. 2015. Genome-wide analysis of Sphingomonas wittichii RW1 behaviour during inoculation and growth in contaminated sand. ISME J 9:150–165. doi: 10.1038/ismej.2014.101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Roggo C, Coronado E, Moreno-Forero SK, Harshman K, Weber J, van der Meer JR. 2013. Genome-wide transposon insertion scanning of environmental survival functions in the polycyclic aromatic hydrocarbon degrading bacterium Sphingomonas wittichii RW1. Environ Microbiol 15:2681–2695. doi: 10.1111/1462-2920.12125 [DOI] [PubMed] [Google Scholar]
- 279. Kota S, Borden RC, Barlaz MA. 1999. Influence of protozoan grazing on contaminant biodegradation. FEMS Microbiol Ecol 29:179–189. doi: 10.1111/j.1574-6941.1999.tb00609.x [DOI] [Google Scholar]
- 280. Otto S, Harms H, Wick LY. 2017. Effects of predation and dispersal on bacterial abundance and contaminant biodegradation. FEMS Microbiol Ecol 93:fiw241. doi: 10.1093/femsec/fiw241 [DOI] [PubMed] [Google Scholar]
- 281. Correa-García S, Pande P, Séguin A, St-Arnaud M, Yergeau E. 2018. Rhizoremediation of petroleum hydrocarbons: a model system for plant microbiome manipulation. Microb Biotechnol 11:819–832. doi: 10.1111/1751-7915.13303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Pizarro-Tobías P, Fernández M, Niqui JL, Solano J, Duque E, Ramos J-L, Roca A. 2015. Restoration of a Mediterranean forest after a fire: bioremediation and rhizoremediation field-scale trial. Microb Biotechnol 8:77–92. doi: 10.1111/1751-7915.12138 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Pizarro-Tobías P, Niqui JL, Roca A, Solano J, Fernández M, Bastida F, García C, Ramos JL. 2015. Field trial on removal of petroleum-hydrocarbon pollutants using a microbial consortium for bioremediation and rhizoremediation. Environ Microbiol Rep 7:85–94. doi: 10.1111/1758-2229.12174 [DOI] [PubMed] [Google Scholar]
- 284. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, Calder PC, Sanders ME. 2014. Expert consensus document. The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 11:506–514. doi: 10.1038/nrgastro.2014.66 [DOI] [PubMed] [Google Scholar]
- 285. Hänninen A, Toivonen R, Pöysti S, Belzer C, Plovier H, Ouwerkerk JP, Emani R, Cani PD, De Vos WM. 2018. Akkermansia muciniphila induces gut microbiota remodelling and controls islet autoimmunity in NOD mice. Gut 67:1445–1453. doi: 10.1136/gutjnl-2017-314508 [DOI] [PubMed] [Google Scholar]
- 286. Cani PD, de Vos WM. 2017. Next-generation beneficial microbes: the case of Akkermansia muciniphila. Front Microbiol 8:1765. doi: 10.3389/fmicb.2017.01765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Costabile A, Bergillos-Meca T, Rasinkangas P, Korpela K, de Vos WM, Gibson GR. 2017. Effects of soluble corn fiber alone or in synbiotic combination with Lactobacillus rhamnosus GG and the pilus-deficient derivative GG-PB12 on fecal microbiota, metabolism, and markers of immune function: a randomized, double-blind, placebo-controlled, crossover study in healthy elderly (Saimes Study). Front Immunol 8:1443. doi: 10.3389/fimmu.2017.01443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Korpela K, Salonen A, Vepsäläinen O, Suomalainen M, Kolmeder C, Varjosalo M, Miettinen S, Kukkonen K, Savilahti E, Kuitunen M, de Vos WM. 2018. Probiotic supplementation restores normal microbiota composition and function in antibiotic-treated and in caesarean-born infants. Microbiome 6:182. doi: 10.1186/s40168-018-0567-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Kumari R, Yadav Y, Misra R, Das U, Das Adhikari U, Malakar P, Dubey GP. 2022. Emerging frontiers of antibiotics use and their impacts on the human gut microbiome. Microbiol Res 263:127127. doi: 10.1016/j.micres.2022.127127 [DOI] [PubMed] [Google Scholar]
- 290. Vatanen T, Ang QY, Siegwald L, Sarker SA, Le Roy CI, Duboux S, Delannoy-Bruno O, Ngom-Bru C, Boulangé CL, Stražar M, Avila-Pacheco J, Deik A, Pierce K, Bullock K, Dennis C, Sultana S, Sayed S, Rahman M, Ahmed T, Modesto M, Mattarelli P, Clish CB, Vlamakis H, Plichta DR, Sakwinska O, Xavier RJ. 2022. A distinct clade of Bifidobacterium longum in the gut of Bangladeshi children thrives during weaning. Cell 185:4280–4297. doi: 10.1016/j.cell.2022.10.011 [DOI] [PubMed] [Google Scholar]
- 291. Douillard FP, de Vos WM. 2019. Biotechnology of health-promoting bacteria. Biotechnol Adv 37:107369. doi: 10.1016/j.biotechadv.2019.03.008 [DOI] [PubMed] [Google Scholar]
- 292. Seegers J, Gül IS, Hofkens S, Brosel S, Schreib G, Brenke J, Donath C, de Vos WM. 2022. Toxicological safety evaluation of live Anaerobutyricum soehngenii strain CH106. J Appl Toxicol 42:244–257. doi: 10.1002/jat.4207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Rodríguez-Daza MC, Pulido-Mateos EC, Lupien-Meilleur J, Guyonnet D, Desjardins Y, Roy D. 2021. Polyphenol-mediated gut microbiota modulation: toward prebiotics and further. Front Nutr 8:689456. doi: 10.3389/fnut.2021.689456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Bertelli C, Pillonel T, Torregrossa A, Prod’hom G, Fischer CJ, Greub G, Giannoni E. 2015. Bifidobacterium longum bacteremia in preterm infants receiving probiotics. Clin Infect Dis 60:924–927. doi: 10.1093/cid/ciu946 [DOI] [PubMed] [Google Scholar]
- 295. Nishiyama K, Yong C-C, Moritoki N, Kitazawa H, Odamaki T, Xiao J-Z, Mukai T, Ercolini D. 2023. Sharing of moonlighting proteins mediates the symbiotic relationship among intestinal commensals. Appl Environ Microbiol 89:e0219022. doi: 10.1128/aem.02190-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Wotzka SY, Nguyen BD, Hardt WD. 2017. Salmonella Typhimurium diarrhea reveals basic principles of enteropathogen infection and disease-promoted DNA exchange. Cell Host Microbe 21:443–454. doi: 10.1016/j.chom.2017.03.009 [DOI] [PubMed] [Google Scholar]
- 297. Ma M, Powell DA, Weyand NJ, Rhodes KA, Rendón MA, Frelinger JA, So M, Payne SM. 2018. A natural mouse model for Neisseria colonization. Infect Immun 86:e00839-17. doi: 10.1128/IAI.00839-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Kirk MD, Pires SM, Black RE, Caipo M, Crump JA, Devleesschauwer B, Döpfer D, Fazil A, Fischer-Walker CL, Hald T, Hall AJ, Keddy KH, Lake RJ, Lanata CF, Torgerson PR, Havelaar AH, Angulo FJ. 2015. World Health Organization estimates of the global and regional disease burden of 22 foodborne bacterial, protozoal, and viral diseases, 2010: a data synthesis. PLoS Med 12:e1001940. doi: 10.1371/journal.pmed.1001940 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. Tsolis RM, Xavier MN, Santos RL, Bäumler AJ, Maurelli AT. 2011. How to become a top model: impact of animal experimentation on human Salmonella disease research. Infect Immun 79:1806–1814. doi: 10.1128/IAI.01369-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300. Unden G, Strecker A, Kleefeld A, Kim OB. 2016. C4-dicarboxylate utilization in aerobic and anaerobic growth. EcoSal Plus 7. doi: 10.1128/ecosalplus.ESP-0021-2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Mølbak K, Simonsen J, Jørgensen CS, Krogfelt KA, Falkenhorst G, Ethelberg S, Takkinen J, Emborg H-D. 2014. Seroincidence of human infections with nontyphoid Salmonella compared with data from public health surveillance and food animals in 13 European countries. Clin Infect Dis 59:1599–1606. doi: 10.1093/cid/ciu627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Favre-Bonté S, Licht TR, Forestier C, Krogfelt KA. 1999. Klebsiella pneumoniae capsule expression is necessary for colonization of large intestines of streptomycin-treated mice. Infect Immun 67:6152–6156. doi: 10.1128/IAI.67.11.6152-6156.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Hutton ML, Mackin KE, Chakravorty A, Lyras D. 2014. Small animal models for the study of Clostridium difficile disease pathogenesis. FEMS Microbiol Lett 352:140–149. doi: 10.1111/1574-6968.12367 [DOI] [PubMed] [Google Scholar]
- 304. Bohnhoff M, Drake BL, Miller CP. 1954. Effect of streptomycin on susceptibility of intestinal tract to experimental Salmonella infection. Proc Soc Exp Biol Med 86:132–137. doi: 10.3181/00379727-86-21030 [DOI] [PubMed] [Google Scholar]
- 305. Litvak Y, Mon KKZ, Nguyen H, Chanthavixay G, Liou M, Velazquez EM, Kutter L, Alcantara MA, Byndloss MX, Tiffany CR, Walker GT, Faber F, Zhu Y, Bronner DN, Byndloss AJ, Tsolis RM, Zhou H, Bäumler AJ. 2019. Commensal Enterobacteriaceae protect against Salmonella colonization through oxygen competition. Cell Host Microbe 25:128–139. doi: 10.1016/j.chom.2018.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306. Eberl C, Weiss AS, Jochum LM, Durai Raj AC, Ring D, Hussain S, Herp S, Meng C, Kleigrewe K, Gigl M, Basic M, Stecher B. 2021. E. coli enhance colonization resistance against Salmonella Typhimurium by competing for galactitol, a context-dependent limiting carbon source. Cell Host Microbe 29:1680–1692. doi: 10.1016/j.chom.2021.09.004 [DOI] [PubMed] [Google Scholar]
- 307. Ruddle SJ, Massis LM, Cutter AC, Monack DM. 2023. Salmonella-liberated dietary L-arabinose promotes expansion in superspreaders. Cell Host Microbe 31:405–417. doi: 10.1016/j.chom.2023.01.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Lentsch V, Woller A, Moresi C, Fattinger SA, Aslani S, Hardt W-D, Loverdo C, Diard M, Slack E. 2022. Combined oral vaccination with niche competition can generate sterilizing immunity against entero-pathogenic bacteria. BioRXiv. doi: 10.1101/2022.07.20.498444 [DOI]
- 309. Raaijmakers JM, Kiers ET. 2022. Rewilding plant microbiomes. Science 378:599–600. doi: 10.1126/science.abn6350 [DOI] [PubMed] [Google Scholar]
- 310. Wilkinson MD, Dumontier M, Aalbersberg IJJ, Appleton G, Axton M, Baak A, Blomberg N, Boiten J-W, da Silva Santos LB, Bourne PE, Bouwman J, Brookes AJ, Clark T, Crosas M, Dillo I, Dumon O, Edmunds S, Evelo CT, Finkers R, Gonzalez-Beltran A, Gray AJG, Groth P, Goble C, Grethe JS, Heringa J, ’t Hoen PAC, Hooft R, Kuhn T, Kok R, Kok J, Lusher SJ, Martone ME, Mons A, Packer AL, Persson B, Rocca-Serra P, Roos M, van Schaik R, Sansone S-A, Schultes E, Sengstag T, Slater T, Strawn G, Swertz MA, Thompson M, van der Lei J, van Mulligen E, Velterop J, Waagmeester A, Wittenburg P, Wolstencroft K, Zhao J, Mons B. 2016. The FAIR guiding principles for scientific data management and stewardship. Sci Data 3:160018. doi: 10.1038/sdata.2016.18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Wood-Charlson EM, Auberry D, Blanco H, Borkum MI, Corilo YE, Davenport KW, Deshpande S, Devarakonda R, Drake M, Duncan WD, Flynn MC, Hays D, Hu B, Huntemann M, Li P-E, Lipton M, Lo C-C, Millard D, Miller K, Piehowski PD, Purvine S, Reddy TBK, Shakya M, Sundaramurthi JC, Vangay P, Wei Y, Wilson BE, Canon S, Chain PSG, Fagnan K, Martin S, McCue LA, Mungall CJ, Mouncey NJ, Maxon ME, Eloe-Fadrosh EA. 2020. The National Microbiome Data Collaborative: enabling microbiome science. Nat Rev Microbiol 18:313–314. doi: 10.1038/s41579-020-0377-0 [DOI] [PubMed] [Google Scholar]
- 312. Pacheco AR, Pauvert C, Kishore D, Segrè D. 2022. Toward FAIR representations of microbial interactions. mSystems 7:e0065922. doi: 10.1128/msystems.00659-22 [DOI] [PMC free article] [PubMed] [Google Scholar]





