Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Seminars in Immunology 66 (2023) 101735

Contents lists available at ScienceDirect

Seminars in Immunology
journal homepage: www.elsevier.com/locate/ysmim

Seminars in immunology special issue: Nutrition, microbiota and immunity


The unexplored microbes in health and disease
Tamar Plitt a, Jeremiah J Faith a, b, *
a
Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
b
Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Functional characterization of the microbiome’s influence on host physiology has been dominated by a few
Gut microbiota characteristic example strains that have been studied in detail. However, the extensive development of methods
Cancer for high-throughput bacterial isolation and culture over the past decade is enabling functional characterization of
In vitro
the broader microbiota that may impact human health. Characterizing the understudied majority of human
High-throughput screen
Gnotobiotics
microbes and expanding our functional understanding of the diversity of the gut microbiota could enable new
Immunology insights into diseases with unknown etiology, provide disease-predictive microbiome signatures, and advance
Culturing microbial therapeutics. We summarize high-throughput culture-dependent platforms for characterizing bacterial
Effector strains strain function and host-interactions. We elaborate on the importance of these technologies in facilitating
mechanistic studies of previously unexplored microbes, highlight new opportunities for large-scale in vitro
screens of host-relevant microbial functions, and discuss the potential translational applications for microbiome
science.

1. Introduction discoveries led to the collecting-of-parts period that we are still in the
early to middle phase of whereby laboratories with advanced methods
Previous decades of research in bacteriology focused on a few model for high-throughput culturing, found approaches to enrich rarer or
organisms and pathogens with a goal of understanding the functions that hard-to-culture microbes, and develop model communities of defined
govern bacterial survival and the factors driving pathogenesis and bacterial strains as consortia for in vivo models or in vitro screens
treatment. Despite this wealth of scientific exploration that included [9–14]. With numerous laboratories now using reference collections of
complete genome sequences [1,2], gene knockout libraries [3], and parts [15–24], the challenge becomes how to identify the functions of so
extensive transcriptional profiling [4], even the best characterized many bacteria when each strain can potentially have a different function
model bacterial strains lack functional annotation of a large proportion from other strains in a species and the number of species alone in the
of their genes [5,6]. human population for gut bacteria is likely in the thousands [25].
In the past 20 years, the microbiome field has transitioned much of Identification of highly penetrant microbial functions in the gut
the bacteriology community’s focus from a few model organisms to­ including short-chain fatty acid (SCFA) production [26], regulatory T
wards the monumentally large challenge of characterizing all the or­ cell (Treg) induction [27], Th17 cell induction [28], and bile acid
ganisms in multiple ecosystems. This change was initially spurred by metabolism [29–33] is already producing functionally important dis­
advances in genome sequencing that enabled a cost-effective means of coveries with translational implications, while fecal microbiota trans­
characterizing the members of bacterial communities using culture- plantation (FMT) and defined live biotherapeutic products (LBPs)
independent approaches [7,8]. Although this cataloging-of-parts phase provide the potential to identify causal influences of bacterial strains in
continues relatively unabated, with technology advances bringing humans, quantify the transmissibility of strains, and translate new
deeper sequencing to identify rarer community members with greater functional discoveries to the clinic. There remains, however, a vast un­
resolution, this phase increased the field’s awareness of the significant explored majority of bacterial strains whose individual and collective
proportion of microbes that were completely unstudied. Such function is largely unknown despite years to centuries of co-habitation

* Corresponding author at: Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
E-mail address: jeremiah.faith@mssm.edu (J.J. Faith).

https://doi.org/10.1016/j.smim.2023.101735
Received 4 October 2022; Received in revised form 17 January 2023; Accepted 9 February 2023
Available online 27 February 2023
1044-5323/© 2023 Elsevier Ltd. All rights reserved.
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

on our bodies. The cataloging-of-parts and the collecting-of-parts periods genomics approaches [23,43,69]. In parallel, larger collections of
of the microbiome field have now set the stage for the new era of defined, sequenced bacterial communities collected into larger consortia
characterization-of-parts whereby we seek high-throughput scalable and screening libraries also provide broad tools for functional explora­
platforms to identify the functions of the understudied majority of tion [15–24]. These collections, inspired by culture-independent dis­
strains in the microbiome. This phase will require studying enough coveries, are now some of the key resources necessary to move from
strains across the phylogenetic tree of life to identify general principles characterizing the functional impact of individual effector strains to
of function across taxa and to identify exceptions that might fuel new understanding the individual and collective role of hundreds to thou­
biological and therapeutic insights. In this review, we summarize sands of strains. We find particularly compelling the isolation and
high-throughput approaches developed to aid in more fully exploring exploration of functional potential of bacterial strains isolated from the
the functional diversity of the human microbiome, with an emphasis on stool of frequently used fecal transplant donors, as these microbes come
the human gut microbiome. We focus on the implications of these from humans that have passed important FMT safety screens. The safety
functional screens for future human translation, particularly on immune data in the resulting recipients provide at least a crude representation of
modulation and cancer. the collective influence of the strains on recipient safety, and the
tracking of strains from donors to recipients provides empirical esti­
2. Generating the microbiome strain catalog mates of the transmissibility of each strain [43].
As the microbiome field transitions from compositional descriptions
2.1. cataloging of parts and phenotypic associations towards studies aimed at mechanistic,
functional, and interventional understanding, there arises an issue of
Culture-independent sequencing transformed our ability to charac­ scale. Every individual is colonized with a unique set of bacterial strains
terize microbial consortia of the human microbiome across body sites which collectively represent enormous functional variation. Purely in
[34]. From the early phase of genus-focused 16S rRNA amplicon vivo based approaches can never approach the scale and the scope
sequencing that identified broad differences between the microbiomes required to address this complexity and so, in vitro assays implemented
of people of different health status and treatment response [35–37], we in high throughput using automation are the only realistic alternative.
have moved to the metagenomics phase of characterizing species-level To tackle questions related to functional responses across microbiota
differences between communities [36,38–42]. Most recently, meta­ derived strains, we can either ignore the complexity and focus on simple
genomics, often combined with strain genome sequencing or meta­ systems (e.g., by using model strains such as segmented filamentous
genomic assembly, is leading to numerous explorations of what defines a bacteria (SFB)) or we must use a variety of tools at different scales to try
strain, how do we track strains [43–47], how many strains exist in a to understand the entire system. The future of human microbial thera­
given species [48] or at a given anatomical location [49], and how peutics will therefore benefit from the parallel advancement of in vitro
strains evolve in their native ecosystems [50–52]. Typically, species are screening platforms, in vivo validation and discovery, and an expansion
defined as isolates that share more than 99% of their 16S rRNA sequence of the transition from FMT interventions [70] to defined bacterial strain
[53,54] while strains are defined as isolates of a given species that share consortia interventions [71–74].
more than 96% of their genome content. [43,48,55,56].
The cataloging of microbiome parts is essential for providing a 2.3. characterization of parts: unknown functions of the understudied
structural framework to build on for functional studies. For example, majority
studying the functional potential of a synthetic human gut microbial
community with mostly Firmicutes and Bacteroides and a few Actino­ Early explorations into the functional potential of human gut
bacteria and Proteobacteria is more realistic to the human gut micro­ microbiota strains, by necessity, focused on a limited number of strains.
biome ecosystem than a community of only Fusobacterium and When there is little to compare to, any significant phenotypic pertur­
Proteobacteria. These microbiome-part-catalogs also provide the vast bation causally linked to a specific bacterial strain is of interest [75] and
majority of functionally insightful microbiome observations directly in many observations ask the simplest question – what is difference be­
humans such as which bacteria engraft in an FMT for subjects with tween a germ-free or antibiotic treated mouse and a mouse colonized
recurrent Clostridium difficile infection [1], ulcerative colitis [57], or with a complex diverse microbiota? Since the goal for a clinical inter­
melanoma [58,59] that respond to FMT therapy. Similarly, enrichments vention will never be to transition someone between a germ-free and
of taxa in specific conditions such as Crohn’s disease [42,60], colorectal colonized state or to provide an antibiotic cocktail to deplete the
cancer [61,62], or wound healing [63] can focus the taxa selection microbiome for life, practical implementation of microbiome functional
process for functional studies and future therapeutics – particularly now discoveries requires identifying individual strains or groups of strains
when more opportunities are emerging for datasets to be combined to that perturb host physiology. The earliest examples of such experiments
search for consensus results across multiple human studies [41,42,60]. identified a few sample strains or consortia that could perturb a host
While the cataloging-of-parts can uncover correlations between the phenotype, for instance, through the induction of Th17 cells [28], Tregs
relative abundance of specific species/strains of bacteria and the [76], or colitis [77] (Fig. 1A). In microbiomes beyond the gut, similar
onset/progression of disease, it is not sufficient to prove causality. experiments identified skin microbiome strains that induce IL-17A+
CD8+ T cells [78] and oral microbes that induce Th1 cells [79]. The
2.2. collecting of parts challenge of these initial forays is that broad conclusions can only be
based on highly significant narrow explorations, and there are far too
Functionally relevant associations from statistical analyses of large few examples to determine how unique the results are relative to all
observational datasets often focus subsequent functional explorations possible experiments with all possible strains.
[64,65]. However, reductionist systems are typically required for broad As modest-sized functional explorations with synthetic defined mi­
functional understanding of biological systems and components. The crobial communities became possible by the advances in gnotobiotic
rise of culture-independent cataloging of the microbiome in health and caging systems and anaerobic culturing, we were able to better under­
disease was closely followed by attempts to culture gut microbial strains stand the context for earlier findings and their potential use in humans
in high-throughput [11], using diverse media and detection technolo­ (Fig. 1B). For example, among gut lamina propria Th17 cells, it appears
gies [13], and with efforts to increase the culturable fraction of the gut that potent Th17 inducing strains [28] are rare strains that are not
[9,10], skin [66], and oral microbiomes [67,68]. Lower cost genome harbored by many people or animals [15,80]. In contrast for Tregs, the
sequencing enabled high-throughput culturing efforts to be coupled microbiome is clearly fundamental to their induction in the lamina
with genome sequencing to facilitate strain tracking and comparative propria [27], and there are a multitude of individual strains and groups

2
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

Fig. 1. Scalable functional characterization of gut microbiome strains. In this illustrative example, the therapeutic aim is to identify bacterial strains that induce high
proliferation in mammalian cells and whose removal or suppression could reduce the risk of colorectal cancer. (A) Early functional explorations of the gut microbiota
focused on a few sample bacterial strains that influenced host physiology. The green dot corresponds to a unique bacteria strain that is present in a healthy donor and
induced low colonic carcinoma cell proliferation. The orange dot corresponds to another unique bacterial strain that is present in a CRC donor and is found to induce
high colonic carcinoma cell proliferation. Both of these bacterial strains belong to the Proteobacteria phylum. Here we might conclude that Proteobacteria in people
with CRC are uniquely capable of inducing colonic carcinoma cell proliferation. (B) Synthetic defined microbial communities enabled exploration of colonic car­
cinoma cell proliferation potential of different bacterial species across multiple taxa per phylum and disease state. Within each disease state, there exist strains that
have a wide range of colonic carcinoma cell proliferative properties. Here we might conclude that multiple phyla contain strains that can induce high colonic
carcinoma cell proliferation, but they are uniquely common in CRC patients. (C) Building on recent expansions of high-throughput culturing, functional screens are
now being performed on the population scale with hundreds of bacterial strains, often with multiple strains per species. On the population scale, we have sufficient
strains in each of our categories of interest (taxonomy and disease status) to see the underlying distributions driving our earlier observations. In this population scale
example, Actinobacteria universally induce high colonic carcinoma cell proliferation and could serve as targets for therapeutic removal or suppression to ultimately
reduced CRC risk, while only a fraction of the Proteobacteria induce high colonic carcinoma cell proliferation. While earlier sample- and consortium-scale exper­
iments showed enrichment of high proliferation-inducing strains in CRC, this effect is explained by CRC enrichment with Actinobacteria and Bacteroides. (D) A
challenge and opportunity for the field moving forward is to develop relevant in vitro and in vivo systems that are the most relevant for advancing microbiome
science towards clinical interventions using the functionally characterized strains. Abbreviation: IBD, inflammatory bowel disease; CRC, colorectal cancer; T1D, type
1 diabetes; FMT, fecal microbiota transplantation.

3
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

of strains able to induce different Treg subsets [76,81,82] through a characterizing the role of the gut microbiota on the gut-brain axis
variety of mechanisms [26,30]. signaling found that metabolites produced by certain strains of Escher­
Empirical observations of strains that repeatedly engraft in fecal ichia coli activated c-Fos in the arcuate nucleus (ARC) and stimulated the
transplants, fractionated fecal transplants, and defined LBPs in subjects firing rate of proopiomelanocortin neurons in the ARC, which play a role
whose disease status is improved by a microbial therapeutic are likely in appetite control and energy expenditure [94,95]. A more recent study
the shortest route to developing minimal bacterial consortia to improve utilized high-coverage metabolomics to describe microbiota-derived
human health. In parallel, there will always be a desire to understand the metabolites implicated in the interorgan transport of gut microbial
mechanisms and general principles driving such successes so that we can metabolites and gut-brain axis [96].
determine the cause of treatment failures, improve future microbial In this review, we will emphasize the role of microbial metabolites in
therapeutics, and envision future applications that are not visible until cancer. Microbial metabolites can have a protective role against carci­
we build the basic foundations necessary to see a little further. To begin nogenesis by contributing to inflammatory tone or influencing the bal­
assembling this foundation as a scientific community will require ance between proliferation and apoptosis in tissues [97]. Several
identifying new in vitro, ex vivo, and in silico methods to bring addi­ microbial metabolites generated from dietary fiber and fats have an
tional scale beyond the animal experiments that have been foundational established effect on cancer [97,98]. Understanding the effects of these
to date and the human observations that are our motivation for explo­ metabolites on cancer growth and spread offers the potential for ther­
ration [70,83]. The microbiome field’s extensive cataloging-of-parts and apeutic interventions – either by direct transfer of strains or adminis­
growing collection-of-parts are the raw resources that were necessary to tration of bacterial products.
open this next era of characterization-of-parts that will facilitate our The intestinal fermentation of dietary fiber releases SCFAs, princi­
understanding of the functional attributes of each strain in a broader pally acetic, propionic, and butyric acids, which help maintain the
context of populations of hundreds of strains (Fig. 1C). integrity of intestinal epithelium [99] and are otherwise unavailable
Here we provide examples from this newly emerging literature in the from diet. These SCFAs have been studied in the context of intratumoral
microbiome field of scientists exploring dozens to hundreds of strains inflammation and demonstrated to have anti-inflammatory effects on
with in vitro, ex vivo, and in silico approaches. For example, larger myeloid cells [100] and colonic Tregs [101–103]. Butyrate, in partic­
explorations, facilitated by ex vivo screens that are less limited by the ular, is the principal energy source for colonic epithelial cells and has
scale of mouse experimentation, suggest that high proportions of Th17 been shown to have tumor suppressive functions [103]. Butyrate can
cells in gnotobiotic mice can be obtained by single potent effector strains inhibit histone deacetylase activity in colonocytes and immune cells,
with highly penetrant phenotype like SFB [28] and similarly high Th17 downregulating proinflammatory cytokines and inducing apoptosis in
cells can be obtained by altering microbiome composition so that it is colorectal cancer (CRC) cells [103–106]. The effects of SCFAs on colonic
composed of only moderate Th17-inducing strains and no low epithelial cells can also be calibrated by the receptors they bind, such as
Th17-inducing strains [84]. We anticipate this emerging trend will G protein-coupled receptors (GPCRs) [107–109]. Gpr109, for instance,
continue to expand as new assays are imagined and more laboratories is expressed on colonic epithelial cells and intestinal myeloid cells and is
have access to large cultured communities of strains. important for Treg generation [109]. Loss of this receptor increases
Ultimately, it will be important to weigh the utility of in vitro, ex susceptibility to colitis associated colorectal cancer (caCRC) [109].
vivo, in vivo, human observational, and human interventional ap­ Propionate activation of Gpr43, expressed in hematopoietic tissues and
proaches in an unbiased manner to identify the best usage of resources in the colon, reduces murine pro-B BaF3 cell proliferation in a
and time to maximize the human relevance of the microbiome field. A dose-dependent manner [110]. Compared to normal colon tissues,
continuous learning and cycling between all of these approaches Gpr43 expression is significantly reduced or lost in colorectal adeno­
(Fig. 1D) is likely necessary for us to reach the point of identifying carcinoma tissues and their corresponding lymph node metastatic ade­
minimal therapeutic sets of strains, strain metabolites, or strain proteins nocarcinomas [106]. Restored Gpr43 expression in human colonic
that cause, prevent, or treat human disease. adenocarcinoma cells led to increased apoptotic cell death upon SCFA
treatment [106].
2.4. Metabolic potential However, conflicting effects of SCFA in cancer prevention have also
been documented. Two mouse studies reported opposing effects of
Screens to understand the metabolic potential of gut microbiota butyrate on tumorigenesis. For instance, butyrate had tumor-promoting
strains and communities represent an area ripe for discovery with major effects in a mouse model of intestinal tumorigenesis driven by mutations
advantages over other types of host/microbe screens. First, the meta­ in the DNA mismatch repair gene, Msh2, and the tumor suppressor gene,
bolic potential of a bacterial strain in vitro and in vivo is likely similar Apc [111]. On the other hand, butyrate ultimately suppressed tumori­
when given the same substrates [22]. Second, there are numerous genesis in the AOM/DSS model of colitis-associated cancer by reducing
human relevant microbiota influenced metabolites to explore from cellular proliferation and increasing apoptosis [92]. The latter effects
SCFAs [85] to drug metabolism [86,87]. In addition, these approaches were explained by the fact that neoplastic colonocytes favor glycolysis
can be performed with either native microbes or in a cloned expression for cellular energy, leading to a decreased butyrate metabolism,
library to facilitate identifying causal genes, metabolites, and enzymes increased nuclear accumulation, and subsequent inhibition of histone
[88,89]. However, challenges and opportunities to the broader appli­ deacetylases. These studies highlight the wide range of effects that a
cation of microbial strain metabolic screens to therapeutics include the single microbial metabolite can elicit in tumor models, hindering the
difficulty of identifying metabolites, and the challenge of comparing translation of microbiome and diet data into practical clinical in­
results across metabolomics measurement platforms. Additionally, the terventions in the context of cancer. Thus, expanding research on the
substantial cross-feeding and metabolic redundancy that occurs in uncharacterized taxa and metabolites will allow us to identify metabo­
multi-species communities can limit the predictability of single strain in lites with more specific or complementary effects.
vitro screens for metabolic processes and compounds for more common Beyond SCFAs, high-fat diet (HFD) and obesity are also thought to be
chemicals in the gut like bile salts and SCFAs. involved in the pathogenesis of specific cancers by altering the gut
The gut microbiota plays a considerable role in host metabolism, microbiota and inducing inflammation [83,98–103,112,114–117]. In
producing active catecholamines [90], regulating serotonin biosynthesis fact, obesity is now regarded as an inflammatory state [97,118] and
[91], and generating butyrate [92]. In fact, evidence suggests that a inflammation and HFD are considered primary risk factors for hepato­
significant proportion of microbial metabolites can translocate from gut cellular carcinoma (HCC) progression [119]. Studies in obese mice
lumen to the systemic circulation influencing distant organs and the treated with an antibiotic cocktail that reduces the number of
central nervous system (CNS) [93]. For instance, studies focused on commensal intestinal bacteria showed a reduced risk of developing HCC

4
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

by depleting the microbiome and the subsequent secretion of Combining in vitro metabolic screens with in vivo validation studies
pro-carcinogenic and proinflammatory metabolites and toxins [120]. already has multiple proof-of-principle demonstrations and appears to
Another mechanism by which HFD influences cancer risk is through be an area that is primed for growth in the next decade. For near term
the secretion of bile acids [97,121]. Primary bile acids are synthesized human relevance, a key will be identifying rare metabolites from gut
from cholesterol in the liver and secreted from the gall bladder into microbial strains that have important beneficial health properties and
duodenum to solubilize and digest fats. Bacteria in the distant small are absent in people with a specific disease. Moving to applications
intestine and colon can metabolize primary bile acids into various sec­ beyond singularly potent metabolite generation or drug metabolism will
ondary bile acids, which are then absorbed in the colon or returned to require understanding the complex problem of what determines the
the liver. Secondary bile acids have been shown to exert immunoregu­ average metabolic output of mixtures of strains with highly overlapping
latory functions and have been implicated in the development and metabolic potential (e.g., short chain fatty acids or secondary bile salt)
progression of liver cancers [121,122]. For instance, in 2018, Ma et al. when given diverse inputs from human diet and host metabolites.
showed that production of secondary bile acids by Clostridium species
contributed to tumor progression in various mouse models of primary or 2.5. Immunogenic potential
metastatic liver cancer [122]. The pro-tumorigenic effect was due to the
secondary bile acid-driven downregulation of CXCL16 in liver sinusoidal 2.5.1. cell mediated immunity
endothelial cells, resulting in a reduced recruitment of natural killer T Given the interconnectivity of the cells in the human immune sys­
(NKT) cells in the liver [122]. In another study, enrichment of certain tem, developing scalable models of immune function is a challenge.
Clostridium species in response to a HFD led to increased levels of the With numerous important roles identified for the microbiota in the
secondary bile acid, deoxycholic acid (DCA), a gut bacterial metabolite shaping of our immune system, it is exciting that large-scale applications
known to cause DNA damage. Accumulation of DCA in the liver led to an are beginning to emerge that take advantage of in vitro, ex vivo, and in
accelerated progression of carcinogen-induced liver cancer [113]. In silico platforms as well as tools from genomics and DNA synthesis.
addition to diet, an earlier study by Yoshimoto found that genetic sus­ Mounting evidence indicates that the microbiome may influence a
ceptibility to obesity can also increase DCA-mediated activation of myriad of immune mediated diseases including inflammatory bowel
mitogenic and proinflammatory response program in hepatic stellate disease (IBD), rheumatoid arthritis, type 2 diabetes, allergic diseases,
cells (HSCs), potentiating liver cancer in mice [120]. and cancers [128]. These data are further supported by studies
Given that the gut microbiota generate, modify, or influence these demonstrating that the gut microbiota directly modulates an antigen
host-relevant metabolites, numerous opportunities exist to broadly specific host immune response [129–131]. For example, Perez-Munoz
characterize the metabolic capacities of bacterial strains in a high- et al. colonized germ-free mice with a strain of Bacteroides thetaiotao­
throughput manner. Recently, Han et.al., demonstrated a metab­ micron and were able to isolate an antigen specific T cell towards the
olomics pipeline to identify and characterize microbiota-dependent strain as well as identify the protein epitope that was shared across
metabolites (MDM) [123]. In a scalable in vitro platform, the team species which was largely predictive of the cross reactivity of the T cell
grew 178 individual human gut microorganisms, spanning 130 species [132]. Yang et al., demonstrated that intestinal Th17 cells in
and 6 phyla, in rich complex culture media and used mass-spectrometry SFB-colonized mice express T-cell antigen receptor (TCR) specific for
to generate a reference dataset of metabolomic profiles for each strain. SFB [133]. Similarly, tolerance to the pathobiont, Helicobacter hepaticus
This screen demonstrated that the phylogenetic relationship between (Hh), was shown to be mediated by Hh-specific Tregs [134]. Treg in­
strains can predict some of the metabolic potential of each strain, with a duction by a range of individual or groups of microbes have been re­
number of exceptions where significant differences exist between ported by multiple groups [27,81,135]. Others have shown the
metabolomic profiles of highly phylogenetically related strains. Scaling specificity of IL17A+ CD8 + T cell response to the skin commensal
this type of platform across multiple growth media and larger numbers Staphylococcus epidermidis lasts for months and is not associated with
of strains will continue to advance our knowledge of the shared and inflammation but rather induced to calibrate barrier immunity and
unique metabolite potential of strains across taxonomy and human provide heterologous protection against invasive pathogens [78].
disease. And ultimately, it may allow us to use comparative genomics to The specificity of the mucosal immune response to the gut com­
associate genomic features with metabolic outputs [123]. In related mensals has profound implications for our understanding of tissue-
work, Wong et al. used a strain-dropout strategy to show that there is specific immunity and pathologies. In accordance with this idea, Spin­
functional redundancy within the bile acid 7a-dehydroxylation niche as dler et al. recently profiled antigen-specific responses directed towards
long as Clostridium scindens or Clostridium hylemonae is present in the individual strains in the setting of a multi-strain defined community and
community [124]. However, dropping out either strain led to variations found lamina propria and mesenteric lymph node T cells specific for
in the bile acid metabolism outside the niche. These studies redefine bacterial strains at the species level, with the ability of Th17 cells to
how we think about strain-level variation in the microbiome. discriminate between some strains of the same species [84]. This spec­
Gut microorganisms are functionally differentiated by their ability to ificity of T cell responses enabled ex vivo recall assays to demonstrate
metabolize specific structurally distinct carbon sources or glycans. the in vivo Th17 contribution of individuals strains in a multi-strain
Synthetic glycans (SGs) from diverse, naturally occurring mono­ community. In addition, knowing the Th17 inducing potential of these
saccharides, or natural glycans that selectively drive the gut micro­ strains allowed for the generation of custom defined strain subsets that
biota’s generation of beneficial metabolites or the relative abundance of collectively induced predictable numbers of Th17 cells in gnotobiotic
specific strains provide one potential therapeutic strategy [125]. Using a mice.
bead-based approach combined with 16S rRNA sequencing, Patnode et. In an important recent application of a scalable in vitro pipeline
al., reported strain-specific and glycan-specific binding phenotypes designed to understand interactions between T cells and gut microbes,
among 160 Bacteroides and Parabacteroides strains [126]. In a more Nagashima et al. colonized germ-free mice with 97 bacterial strains to
recent screen of carbohydrate utilization of 354 members across 29 educate their immune system with the multi-strain consortia [69].
species of the Bacteroidetes phylum [127], Pudlo et al. found strongly Two-weeks later, T cells from the large intestine were co-cultured ex
enriched glycan utilization abilities within species demonstrating a vivo with each of the 97 strains to identify the specificity of the T cells
vertical evolution functional imprint on gut microbial strains. However, towards each strain. Although the global T cell pool was clearly being
similar to studies by Han et al., a clear mosaicism existed at the level of driven by antigen-specific T cells towards certain strains, it was also
individual strains suggesting that horizontal gene transfer or perhaps found that many T cells recognized multiple strains of bacteria. Using a
convergent evolution provide important contributions to the metabolic beautiful combination of TCR-sequencing to identify relevant T cells,
capacity of individual strains. DNA synthesis, a T cell reporter line to generate 92 T cell clonotypes,

5
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

and comparative genomics across the large strain library, the authors microbiota [142]. Colonization of GF mice with bacteria from human
identified abundant TCR clonotypes that were polyspecific for 18 Fir­ IBD patients that were highly coated with IgA conferred dramatic sus­
micutes in the strain collection. In addition, the authors discovered that ceptibility to colitis. In addition, Alexander et al. characterized antibody
these 18 strains shared a conserved substrate binding protein from an reactivity in IBD patients using microbiota antigen arrays directed at
ABC transport system that turned out to be a T cell epitope. In many recombinant bacterial flagellins [143]. Compared to healthy controls, a
ways, these observations parallel prior work with small numbers of subset of patients with Crohn’s disease displayed elevated serum IgG
strains that identified a specific T cell epitope to B. thetaiotaomicron and response to multiple flagellins. The multi-flagellin reactive patients also
the shared reactivity of an antigen specific T cell towards other members exhibited elevated flagellin-specific T effector memory (TEM) cells, a
of the Bacteroidetes [132]. However, by taking advantage of more reduced ratio of flagellin-reactive Treg to effector T cells, and a high
scalable platforms and larger communities of bacterial strains, we have frequency of disease complications. Understanding the specificity of
the opportunity to pursue more interactions and to have a better un­ antibodies towards gut microbial strains is an area that is well suited for
derstanding of how such observations might extrapolate to humans high-throughput functional screening experiments. The availability of
colonized with large numbers of microbes in an ecosystem that may gain strain- and species-specific hybridomas along with the numerous
or lose strains over time. antibody-based in vitro assays currently in the literature facilitate
High-throughput screening platforms have also been developed for high-throughput screens.
identifying interactions between microbes and innate immune cells. For In addition to IgA, intestinal IgG and IgM antibodies have also been
example, a recent screen of a synthetic plant root bacterial community reported to react to the gut commensal antigens [144]. While these
identified strains that suppress MAMP-triggered immunity induced by antibodies are not as well-characterized, they share similar specificities
flg2 (a 22-amino acid epitope in bacterial flagellin). This immune sup­ to IgA. Homeostatic commensal IgG antibodies, specifically IgG2b and
pression influenced the ability of different bacteria to colonize the root IgG3, commonly bind gram-negative antigens (i.e., Proteobacteria).
and could be manipulated with subsets of the 35 strains identified to IgG2b and IgG3 are transmitted to neonates in breast milk and maternal
have suppressive effects in a monocolonization screen [136]. Another transmission of these IgG antibodies coordinates with IgA to limit
recent study explored the interactions between gut microbial strains and mucosal T cell responses, and modulates neonatal intestinal ILC3 and
toll like receptors (TLRs). TLRs are a major sub-family of pattern macrophage differentiation [146]. Zeng et al., identified murein lipo­
recognition receptors (PRRs) that are widely expressed by a variety of protein (MLP) as a major antigen that induced systemic IgG in both mice
cells. In this study, 277 commensal bacterial strains from healthy in­ and humans [145]. Administration of anti-MLP conferred protection
dividuals and individuals with IBD were co-cultured with myeloid cells against Salmonella infection.
or dendritic cells to quantify the cytokine response to each strain [23].
The cytokine responses to these 277 gut bacterial strains were found to 3. Oncogenic potential
be as strong as responses toward enteric pathogens and varied across
phyla, down to the strain level [23]. Similar to the population scale Perturbations in the composition and metabolic output of the gut
metabolomics screen [123] and the carbohydrate utilization profiles microbiota are associated with diseases ranging from autoimmune and
from a large screening library [127], a great deal of phylogenetically metabolic diseases to cancer. The mechanisms by which microbes
enriched patterns could be identified in the innate immune responses contribute to carcinogenesis fall into three broad categories: 1) pro­
towards gut bacterial strains, while numerous exceptions to these phy­ moting gastrointestinal (GI) inflammation, 2) inducing genotoxicity
logenic patterns highlight how strain diversity and horizontal gene within intestinal epithelial cells, and 3) promoting cell proliferation of
transfer allows for a great diversity of responses. Moreover, Spindler intestinal epithelial cells [97].
et al. found that myeloid cells differentially rely on toll-like receptor 2
(TLR2) or TLR4 activation to sense particular taxa, an observation that 3.1. Inflammation
predicted in vivo function. Finally, in vitro co-culture assays have also
been successfully employed to explore the functional potential of strains Although mucosal barriers maintain symbiotic relationship between
from the cervicovaginal microbiome on the cervicovaginal epithelial the gut microbiota and the host, they are susceptible to constant envi­
barrier [137] and represent one of the more generalizable and scalable ronmental insults. Cancer and inflammatory disorders arise when these
systems for exploring other microbiomes. mucosal surface barriers are breached, and microbes pass into the
lamina propria, overturning immune tolerance and inciting mucosal
2.6. Humoral immunity inflammation [97,147–149]. Upon mucosal barrier breach, proin­
flammatory pathways become activated serving as a critical component
IgA responses have been demonstrated to be influenced by the gut of tumor progression. Inflammatory factors in the tumor microenvi­
microbiome [138–141]. Yang et al. evaluated fecal IgA responses to ronment, such as reactive oxygen species (ROS) and reactive nitrogen
commensal bacteria and found Bacteroides ovatus to be the most prom­ species (RNS), further contribute to tumor growth and metastasis [150].
inent IgA inducer in the colon of gnotobiotic mice [139]. Further Subsequent tumor engagement of PRRs results in further cell prolifera­
characterization of 19 unique B. ovatus strains revealed strain-specific tion by activating cell survival signaling through NF-kB, a master
IgA responses. Transplanting low-IgA mice with a multiplex cocktail regulator in cancer-associated inflammation [151–153]. For instance,
of high-IgA inducing strains, but not individual ones, led to an increase CRC-associated Fusobacterium nucleatum has been shown to activate
of fecal IgA. Follow up studies found the bulk secretory IgA antibody NF-kB signaling within the tumor microenvironment through various
repertoire tailored to recognize the “self” gut bacteria with a minor mechanisms, such as PRR engagement [154–156] or FadA binding of
fraction of IgAs displaying cross-reactivity [24]. At the same time, E-cadherin on epithelial cells [157]. NF-kB regulates the transcription of
Rollenske et al. identified a range of antigen-specific monoclonal IgAs over 100 proinflammatory, as well as anti-apoptotic, genes. Nod-like
with a diverse immunoglobulin gene repertoire targeting defined bac­ receptors (NLR), which are cytosolic PRRs, have also been linked to
terial antigens [138]. The distinct functional effects of these IgAs were cancer as mice deficient in NLRs display an enhanced susceptibility to
highly dependent on the antigen recognized. caCRC [158–161]. For instance, colons of NOD2-/- mice express signif­
In the context of disease, multiple groups found taxa specific IgA icantly higher levels of inflammatory genes and activation of inflam­
responses in relation to perturbations in the gut microbiota. For matory pathways [158].
instance, Palm et al. examined IgA coating patterns in mice with a Beyond the impact on the innate immune system, mucosal barrier
transmissible colitogenic microbiota and found high IgA coating spe­ breach also has deleterious consequences on the adaptive immune arm.
cifically marked known disease-driving members of the intestinal The inappropriate interaction between intestinal epithelium, gut

6
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

microbiota, and the immune cells is the major contributor to inflam­ exposed to lysates from CDT-deficient C. jejuni [178].
matory pathogenesis of the gut. Numerous studies have implicated the While insofar these genotoxic assays of cancer-associated microbes
role of the IL23/IL17 axis [162,163], TNFa [152,164,165], T follicular described above remain limited to a handful of bacterial strains, it re­
helper cells, [166] and STAT3 [167] in tumor growth and progression. mains possible to scale up these assays to evaluate and predict the
As discussed above, several in vitro and ex vivo screening platforms genotoxic (and potentially, carcinogenic) potential of additional strains.
have been developed for identifying interactions between hundreds of In an excellent recent example of such a scale-up, Cao et al., screened the
microbes and immune cells including the innate and adaptive cells genotoxicity of more than 100 phylogenetically diverse human gut
associated with cancer. The output from such screens will be important commensal bacterial strains through co-incubation of each strain with
in understanding the inflammatory contributions of bacterial strains’ plasmid DNA followed by gel electrophoresis [188]. They identified
oncogenic potential. several new genotoxic strains, a subset of which drove increased tumor
formation in susceptible mice, and the genotoxic metabolite produced
3.2. Genotoxicity by one of their top hits – a strain of Morganella morganii.

Beyond eliciting inflammation, many bacteria have evolved mech­ 3.3. Proliferation
anisms to damage DNA to confer a survival advantage over other mi­
croorganisms [168,169]. These mechanisms are important for bacterial Several cancer-associated bacteria were shown to alter signaling
ecology and evolution, with evidence that such behavior can prevent pathways involved in cell stemness and growth. The Wnt/β-catenin
competing strains from invading a niche or inhibit the growth of coex­ pathway is a pleiotropic signaling pathway commonly mutated in many
isting strains [168,170]. However, these bacterial defense strategies can cancers. This pathway regulates embryo development, cell proliferation
lead to host chromosomal instability - a hallmark of carcinogenesis. For and differentiation, and apoptosis [189]. Unsurprisingly, pathogenic
instance, E. coli (as well as other Enterobacteriaceae) expressing the bacteria have evolved strategies to hijack these processes to establish a
genomic island polyketide synthase (pks+) produces the genotoxin col­ replicative niche [190]. Bacteria such as Helicobacter pylori, F. nucleatum,
ibactin that is believed to alkylate adenine residues in DNA, causing ETBF, Salmonella typhi, Mycobacterium tuberculosis, and C. difficile secrete
DNA damage [171,172]. The detection of pks+ E. coli in human CRCs, virulence factors that alter the activity of the Wnt/β-catenin pathway
along with the ability of these strains to potentiate tumorigenesis in [97,189]. For instance, oncogenic strains of H. pylori inject an exogenous
animal models of disease, generated great interest in colibactin as a cancer-promoting protein, CagA, intermittently into gastric epithelial
molecule of interest in colorectal carcinogenesis [173–176]. Cytolethal cells leading to activation of the Wnt/β-catenin pathway and subsequent
distending toxin (CDT) produced by several ε- and γ-proteobacteria has upregulation of genes involved in proliferation, survival, migration, and
also been shown to produce DNA double-strand breaks (DSBs) in angiogenesis [97,151,191–193]. On the other hand, F. nucleatum, a
mammalian cells and promote colorectal tumorigenesis in mice [177, member of the oral microbiota that is associated with human colorectal
178]. Accumulating data also support the role for enterotoxigenic Bac­ adenomas, uses its FadA adhesin to bind and invade epithelial and
teroides fragilis (ETBF), Enterococcus faecalis, and Peptostreptococcus endothelial cells [157,194–196]. FadA binds host E-cadherin, activating
anaerobius in animal and human tumorigenesis via ROS-mediated the Wnt/β-catenin pathway and stimulating the growth of cancer cells
damage of host DNA [179–182]. At high levels, ROS can lead to DNA [157]. ETBF, enriched in some human CRCs, secretes a 20-kDa metal­
damage and mutations. loprotease toxin, BFT [197–199]. This toxin cleaves host E-cadherin,
The characterization and measurement of genotoxicity may be leading to the stimulation of β-catenin-dependent cell proliferation
extremely valuable in predicting the pathogenicity of bacterial strains, [198,200]. Finally, S. typhi, overabundant in hepatobiliary cancers,
specifically those associated with increased cancer risk. Many of the activate the β-catenin pathway through its pathogenic product, AvrA
traditional DNA damage test methods, such as the bacterial reverse [201–203]. AvrA alters ubiquitination and acetylation of target proteins
mutation (Ames) test, are laborious, time-consuming and harbor a va­ to modulate epithelial proliferation and apoptosis [203,204].
riety of other methodological issues including low sample throughput Proliferation is an important part of cancer development and pro­
[183,184]. However, a number of sensitive, high-throughput genotox­ gression and so, screening bacterial strains for their ability to alter
icity platforms have been recently developed for rapid measurement of mammalian cell proliferation could identify potentially oncogenic mi­
DNA damage (single- and double-strand breaks). Most notably, the crobes. For instance, CRC-associated ETBF has been shown to stimulate
extent of DNA damage in intestinal epithelial cells has been quantified in cell proliferation through the secretion of BFT in multiple cancerous
vitro and in vivo using phosphorylated histone H2AX (γH2AX) as a intestinal epithelial cell lines using the thymidine incorporation assay
surrogate marker of DNA damage. Immunofluorescence (IF) staining of [200]. Furthermore, immunohistochemistry evaluation of β-catenin
rat intestinal epithelial cells (IEC-6) infected with the pks+ harboring abundance in the colon of mice colonized with Citrobacter rodentium
strain of E. coli NC101 revealed γH2AX in ~30% of cells compared with revealed colonocyte hyperproliferation [205]. C. rodentium has been
γH2AX in < 5% in cells infected with isogenic pks-deficient NC101 shown to promote adenoma formation in ApcMin/+ mice [206]. Finally,
[175]. Similar results have been reported in germ-free IL10-/- mice and BrdU and XTT proliferation assays have been used to show proliferation
transparent zebrafish larva using immunohistochemistry (IHC) and and migration of pancreatic cancer cells upon F. nucleatum invasion
flow-cytometry based detection assays of γH2AX [185]. Smaller-scale IF [207]. Many of these proliferation assays can be modified to scale up the
screens using an antibody against the DNA lesion 8-oxo-7,8-dihy­ screening of pro-proliferative microbes associated with various diseases.
dro-2′ -deoxyguanosine (8-oxoG), a more general marker of oxidative
DNA damage, have also been used to demonstrate the effects of microbe 4. Challenges and opportunities
induced DNA damage in IL10-/- inflammatory CRC models [186].
The mechanism of CDT-induced DNA damage was defined using Performing functional screens at the scale of hundreds to thousands
similar, although low-throughput, genotoxic screens including IF using of microbial strains provides both advantages and disadvantages over
antibodies against a well-defined DSB markers 53BP [187] and the more common experiments on individual strains or small groups of
comet assay [178], which is another reliable method for measuring DNA strains. To practically move beyond one-hundred different strains
strand breaks. Bacterial lysates from wild-type and CDT-deficient assayed individually requires a break from individual gnotobiotic mice
Campylobacter jejuni strains were used to treat the human colon cancer and a transition to either ex vivo assays on mice previously colonized
cell line HT-29, IEC-6 cell line, and culture enteroids for 24 h. In all three with sets of strains or in vitro assays. A key feature of these large-scale
experimental models, bacterial lysates from wild-type C. jejuni promoted platforms will then be the relevance of the implemented assay for in
DNA damage, while this response was strongly attenuated in cells vivo and human biology. Furthermore, working with hundreds of

7
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

microbial strains is inherently more error prone than working with small [3] T. Baba, et al., Construction of Escherichia coli K-12 in-frame, single-gene
knockout mutants: the Keio collection, Mol. Syst. Biol. 2 (2006) (2006) 0008.
numbers of strains. During such screens, it is not reasonable to assume
[4] J.J. Faith, et al., Large-scale mapping and validation of Escherichia coli
that each strain will grow every time the assay is run and that there will transcriptional regulation from a compendium of expression profiles, PLoS Biol. 5
not be a contamination of any wells if hundreds of strains are being (2007), e8.
grown in multi-well plates in close proximity. Therefore, it is critical that [5] S. Kasif, R.J. Roberts, We need to keep a reproducible trace of facts, predictions,
and hypotheses from gene to function in the era of big data, PLoS Biol. 18 (2020),
the purity of each strain is assayed with each screen to eliminate any e3000999.
wells that are not the expected strain or that did not grow. Furthermore, [6] R.J. Roberts, et al., COMBREX: a project to accelerate the functional annotation
we need improved methods for replicating and distributing screening of prokaryotic genomes, Nucleic Acids Res 39 (2011) D11–D14.
[7] D.J. Lane, et al., Rapid determination of 16S ribosomal RNA sequences for
libraries of bacterial strains so that assays can be repeated across labs phylogenetic analyses, Proc. Natl. Acad. Sci. USA 82 (1985) 6955–6959.
and different screens can be performed on the same sets of strains [8] C.R. Woese, G.E. Fox, Phylogenetic structure of the prokaryotic domain: the
allowing for a growing knowledgebase of these strains. primary kingdoms, Proc. Natl. Acad. Sci. USA 74 (1977) 5088–5090.
[9] H.P. Browne, et al., Culturing of ‘unculturable’ human microbiota reveals novel
Other challenges that lie ahead for large-scale functional screens of taxa and extensive sporulation, Nature 533 (2016) 543–546.
microbiome strains include how to build on prior knowledge rather than [10] M. Derrien, E.E. Vaughan, C.M. Plugge, W.M. de Vos, Akkermansia muciniphila
have each study be an island. Our accumulation of knowledge from the gen. nov., sp. nov., a human intestinal mucin-degrading bacterium, Int J. Syst.
Evol. Microbiol 54 (2004) 1469–1476.
scientific community functional efforts have traditionally been focused [11] A.L. Goodman, et al., Extensive personal human gut microbiota culture
across individual bacterial strains, model organisms (e.g., mouse), or collections characterized and manipulated in gnotobiotic mice, Proc. Natl. Acad.
human. Heavily used human functional resources like TCGA [208] and Sci. USA 108 (2011) 6252–6257.
[12] E.E. Hansen, et al., Pan-genome of the dominant human gut-associated archaeon,
ImmGen [209] have an advantage that they target a single or limited
Methanobrevibacter smithii, studied in twins, Proc. Natl. Acad. Sci. USA 108
number of mammalian hosts, whereas in the context the human (Suppl 1) (2011) 4599–4606.
microbiome, each new screen could include hundreds of new strains that [13] J.-C. Lagier, et al., Microbial culturomics: paradigm shift in the human gut
have never been studied before. At the same time, there is high overlap microbiome study, Clin. Microbiol Infect. 18 (2012) 1185–1193.
[14] F.E. Rey, et al., Metabolic niche of a prominent sulfate-reducing human gut
at the species and other taxonomic levels and even in the genetic se­ bacterium, Proc. Natl. Acad. Sci. USA 110 (2013) 13582–13587.
quences available for more strains used in high-throughput screens. [15] G.J. Britton, et al., Defined microbiota transplant restores Th17/RORγt(+)
Therefore, envisioning systems that organize the results from different regulatory T cell balance in mice colonized with inflammatory bowel disease
microbiotas, Proc. Natl. Acad. Sci. USA 117 (2020) 21536–21545.
high-throughput screens into a shared community database is a daunt­ [16] S. Brugiroux, et al., Genome-guided design of a defined mouse microbiota that
ing but not impossible task – one that requires some ingenuity to confers colonization resistance against Salmonella enterica serovar Typhimurium,
maximize the utility across studies but with a potential payback of Nat. Microbiol 2 (2016) 16215.
[17] A.G. Cheng, et al., Design, construction, and in vivo augmentation of a complex
increased data reutilization, a growing knowledge of the probability of gut microbiome, Cell 185 (3617–3636) (2022), e19.
different functions as a function of taxonomic or genetic context, and [18] C.S. Eun, et al., Induction of bacterial antigen-specific colitis by a simplified
community organization. Other key aspects of functional screens human microbiota consortium in gnotobiotic interleukin-10-/- mice, Infect.
Immun. 82 (2014) 2239–2246.
include the scale of the screen and the generalizability of results. With [19] J.J. Faith, et al., Creating and characterizing communities of human gut microbes
dozens of organisms one can cover multiple common species, with in gnotobiotic mice, ISME J. 4 (2010) 1094–1098.
hundreds of organisms more rare species will be observed as well as [20] S.C. Forster, et al., A human gut bacterial genome and culture collection for
improved metagenomic analyses, Nat. Biotechnol. 37 (2019) 186–192.
multiple strains of common species, and with thousands of organisms in
[21] M. Poyet, et al., A library of human gut bacterial isolates paired with longitudinal
a screen multiple strains can be tested for each species. As more genomes multiomics data enables mechanistic microbiome research, Nat. Med 25 (2019)
become available for each bacterial species, pangenomics will also 1442–1452.
provide a strategy to select strains and determine the number of strains [22] K.A. Romano, E.I. Vivas, D. Amador-Noguez, F.E. Rey, Intestinal microbiota
composition modulates choline bioavailability from diet and accumulation of the
necessary to cover a specified fraction of a species’ pangenomic space. proatherogenic metabolite trimethylamine-N-oxide, mBio 6 (2015), e02481.
With the majority of high-throughput functional screens of over 100 [23] M.P. Spindler, et al., Human gut microbiota stimulate defined innate immune
microbial strains emerging as publications or preprints only in the past responses that vary from phylum to strain, 00408–5, Cell Host Microbe
S1931–3128 (22) (2022), https://doi.org/10.1016/j.chom.2022.08.009.
year, it is likely that we will see this trend expanding in the coming [24] C. Yang, et al., Immunoglobulin A antibody composition is sculpted to bind the
years. As a community, we must balance the utility of each lab using self gut microbiome, Sci. Immunol. 7 (2022), eabg3208.
different screening sets of strains versus shared strains across screens [25] J. Qin, et al., A human gut microbial gene catalogue established by metagenomic
sequencing, Nature 464 (2010) 59–65.
versus some combination of the two. Regardless, these larger screens [26] P.M. Smith, et al., The microbial metabolites, short-chain fatty acids, regulate
that often include multiple strains per species for common species pro­ colonic Treg cell homeostasis, Science 341 (2013) 569–573.
vide a wealth of information across studies even when different strains [27] K. Atarashi, et al., Treg induction by a rationally selected mixture of Clostridia
strains from the human microbiota, Nature 500 (2013) 232–236.
are used because the higher-level trends observed across taxonomic [28] I.I. Ivanov, et al., Induction of intestinal Th17 cells by segmented filamentous
groups are easily transferable to other strains. The key to success for bacteria, Cell 139 (2009) 485–498.
these new types of experiments will be to balance the value of the [29] C.G. Buffie, et al., Precision microbiome reconstitution restores bile acid
mediated resistance to Clostridium difficile, Nature 517 (2015) 205–208.
screening assays, in vivo validations, and human interventions to ulti­
[30] C. Campbell, et al., Bacterial metabolism of bile acids promotes generation of
mately determine the utility of new discoveries. peripheral regulatory T cells, Nature 581 (2020) 475–479.
[31] S. Hang, et al., Bile acid metabolites control T(H)17 and T(reg) cell
Acknowledgements differentiation, Nature 576 (2019) 143–148.
[32] L.N. Lucas, et al., Dominant Bacterial Phyla from the Human Gut Show
Widespread Ability To Transform and Conjugate Bile Acids, mSystems e0080521
This work was supported by National Institutes of Health grants (nos. (2021), https://doi.org/10.1128/mSystems.00805-21.
NIDDK DK112978, NIDDK DK124133, and NIDDK DK123749). We [33] D. Paik, et al., Human gut bacteria produce Τ(Н)17-modulating bile acid
metabolites, Nature 603 (2022) 907–912.
thank Graham Britton for critical comments on the manuscript and [34] Structure, function and diversity of the healthy human microbiome, Nature 486
Philip Ahern for helpful tweets on T cell specificity. (2012) 207–214.
[35] P.B. Eckburg, et al., Diversity of the human intestinal microbial flora, Science 308
(2005) 1635–1638.
References [36] D. Gevers, et al., The treatment-naive microbiome in new-onset Crohn’s disease,
Cell Host Microbe 15 (2014) 382–392.
[1] F.R. Blattner, et al., The complete genome sequence of Escherichia coli K-12, [37] J.M. Shapiro, et al., Immunoglobulin a targets a unique subset of the microbiota
Science 277 (1997) 1453–1462. in inflammatory bowel disease, Cell Host Microbe 29 (83–93) (2021), e3.
[2] T. Hayashi, et al., Complete genome sequence of enterohemorrhagic Escherichia [38] E.A. Franzosa, et al., Gut microbiome structure and metabolic activity in
coli O157:H7 and genomic comparison with a laboratory strain K-12, DNA Res 8 inflammatory bowel disease, Nat. Microbiol 4 (2019) 293–305.
(2001) 11–22. [39] V. Gopalakrishnan, et al., Gut microbiome modulates response to anti-PD-1
immunotherapy in melanoma patients, Science 359 (2018) 97–103.

8
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

[40] B. Routy, et al., Gut microbiome influences efficacy of PD-1-based [75] G.J. Britton, J.J. Faith, Causative Microbes in Host-Microbiome Interactions,
immunotherapy against epithelial tumors, Science 359 (2018) 91–97. Annu Rev. Microbiol 75 (2021) 223–242.
[41] F.Y. Shaikh, et al., A uniform computational approach improved on existing [76] M.B. Geuking, et al., Intestinal bacterial colonization induces mutualistic
pipelines to reveal microbiome biomarkers of nonresponse to immune checkpoint regulatory T cell responses, Immunity 34 (2011) 794–806.
inhibitors, Clin. Cancer Res 27 (2021) 2571–2583. [77] H.C. Rath, et al., Normal luminal bacteria, especially Bacteroides species, mediate
[42] B. Yilmaz, et al., Microbial network disturbances in relapsing refractory Crohn’s chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin
disease, Nat. Med 25 (2019) 323–336. transgenic rats, J. Clin. Invest 98 (1996) 945–953.
[43] V. Aggarwala, et al., Precise quantification of bacterial strains after fecal [78] S. Naik, et al., Commensal-dendritic-cell interaction specifies a unique protective
microbiota transplantation delineates long-term engraftment and explains skin immune signature, Nature 520 (2015) 104–108.
outcomes, Nat. Microbiol 6 (2021) 1309–1318. [79] K. Atarashi, et al., Ectopic colonization of oral bacteria in the intestine drives T
[44] M.R. Olm, et al., inStrain profiles population microdiversity from metagenomic (H)1 cell induction and inflammation, Science 358 (2017) 359–365.
data and sensitively detects shared microbial strains, Nat. Biotechnol. 39 (2021) [80] M. Viladomiu, et al., IgA-coated E. coli enriched in Crohn’s disease
727–736. spondyloarthritis promote T(H)17-dependent inflammation, Sci. Transl. Med 9
[45] D.T. Truong, A. Tett, E. Pasolli, C. Huttenhower, N. Segata, Microbial strain-level (2017).
population structure and genetic diversity from metagenomes, Genome Res 27 [81] J.J. Faith, P.P. Ahern, V.K. Ridaura, J. Cheng, J.I. Gordon, Identifying gut
(2017) 626–638. microbe-host phenotype relationships using combinatorial communities in
[46] S. Zhao, C.L. Dai, E.D. Evans, Z. Lu, E.J. Alm, Tracking strains predicts personal gnotobiotic mice, Sci. Transl. Med 6 (2014) 220ra11.
microbiomes and reveals recent adaptive evolution, 2020.09.14, bioRxiv (2020), [82] N. Geva-Zatorsky, et al., Mining the Human Gut Microbiota for
296970, https://doi.org/10.1101/2020.09.14.296970. Immunomodulatory Organisms, Cell 168 (928–943) (2017), e11.
[47] W. Zheng, et al., High-throughput, single-microbe genomics with strain [83] S. Paramsothy, et al., Multidonor intensive faecal microbiota transplantation for
resolution, applied to a human gut microbiome, Science 376 (2022) eabm1483. active ulcerative colitis: a randomised placebo-controlled trial, Lancet 389 (2017)
[48] J.J. Faith, et al., Strain population structure varies widely across bacterial species 1218–1228.
and predicts strain colonization in unrelated individuals, 2020.10.17, bioRxiv [84] M.P. Spindler, I. Mogno, P. Suri, G.J. Britton, J.J. Faith, Species-specific CD4+ T
(2020), 343640, https://doi.org/10.1101/2020.10.17.343640. cells Enable Prediction of Mucosal Immune Phenotypes from Microbiota
[49] A. Conwill, et al., Anatomy promotes neutral coexistence of strains in the human Composition, 2022.08.13, bioRxiv (2022), 503851, https://doi.org/10.1101/
skin microbiome, Cell Host Microbe 30 (171–182) (2022), e7. 2022.08.13.503851.
[50] T.D. Lieberman, Detecting bacterial adaptation within individual microbiomes, [85] D.J. Morrison, T. Preston, Formation of short chain fatty acids by the gut
Philos. Trans. R. Soc. Lond. B Biol. Sci. 377 (2022) 20210243. microbiota and their impact on human metabolism, Gut Microbes 7 (2016)
[51] B. Yilmaz, et al., Long-term evolution and short-term adaptation of microbiota 189–200.
strains and sub-strains in mice, Cell Host Microbe 29 (650–663) (2021), e9. [86] M. Zimmermann, M. Zimmermann-Kogadeeva, R. Wegmann, A.L. Goodman,
[52] S. Zhao, et al., Adaptive evolution within gut microbiomes of healthy people, Cell Mapping human microbiome drug metabolism by gut bacteria and their genes,
Host Microbe 25 (656–667) (2019), e8. Nature 570 (2019) 462–467.
[53] J.S. Johnson, et al., Evaluation of 16S rRNA gene sequencing for species and [87] N. Koppel, V. Maini Rekdal, E.P. Balskus, Chemical transformation of xenobiotics
strain-level microbiome analysis, Nat. Commun. 10 (2019) 5029. by the human gut microbiota, Science 356 (2017).
[54] R.C. Edgar, Updating the 97% identity threshold for 16S ribosomal RNA OTUs, [88] L.J. Cohen, et al., Unraveling function and diversity of bacterial lectins in the
Bioinformatics 34 (2018) 2371–2375. human microbiome, Nat. Commun. 13 (2022) 3101.
[55] A. Chen-Liaw, et al., Gut microbiota bacterial strain richness is species specific [89] D.A. Colosimo, et al., Mapping Interactions of Microbial Metabolites with Human
and limits therapeutic engraftment, 2022.11.01, bioRxiv (2022), 514782, G-Protein-Coupled Receptors, Cell Host Microbe 26 (273–282) (2019), e7.
https://doi.org/10.1101/2022.11.01.514782. [90] Y. Asano, et al., Critical role of gut microbiota in the production of biologically
[56] J.J. Faith, et al., The long-term stability of the human gut microbiota, Science 341 active, free catecholamines in the gut lumen of mice, Am. J. Physiol. Gastrointest.
(2013) 1237439. Liver Physiol. 303 (2012) G1288–G1295.
[57] S. Paramsothy, et al., Specific Bacteria and Metabolites Associated With Response [91] J.M. Yano, et al., Indigenous bacteria from the gut microbiota regulate host
to Fecal Microbiota Transplantation in Patients With Ulcerative Colitis, serotonin biosynthesis, Cell 161 (2015) 264–276.
Gastroenterology 156 (1440–1454) (2019), e2. [92] D.R. Donohoe, et al., A gnotobiotic mouse model demonstrates that dietary fiber
[58] E.N. Baruch, et al., Fecal microbiota transplant promotes response in protects against colorectal tumorigenesis in a microbiota- and butyrate-
immunotherapy-refractory melanoma patients, Science 371 (2021) 602–609. dependent manner, Cancer Disco 4 (2014) 1387–1397.
[59] D. Davar, et al., Fecal microbiota transplant overcomes resistance to anti-PD-1 [93] F. Hosseinkhani, et al., The contribution of gut bacterial metabolites in the human
therapy in melanoma patients, Science 371 (2021) 595–602. immune signaling pathway of non-communicable diseases, Gut Microbes 13
[60] M. Schirmer, A. Garner, H. Vlamakis, R.J. Xavier, Microbial genes and pathways (2021) 1–22.
in inflammatory bowel disease, Nat. Rev. Microbiol 17 (2019) 497–511. [94] N. Tennoune, et al., Bacterial ClpB heat-shock protein, an antigen-mimetic of the
[61] S. Tomkovich, et al., Human colon mucosal biofilms from healthy or colon cancer anorexigenic peptide α-MSH, at the origin of eating disorders, Transl. Psychiatry
hosts are carcinogenic, J. Clin. Invest 129 (2019) 1699–1712. 4 (2014), e458.
[62] F. Colombo, et al., Gut microbiota composition in colorectal cancer patients is [95] J. Breton, et al., Gut Commensal E. coli Proteins Activate Host Satiety Pathways
genetically regulated, Sci. Rep. 12 (2022) 11424. following Nutrient-Induced Bacterial Growth, Cell Metab. 23 (2016) 324–334.
[63] S. Gupta, et al., Cutaneous surgical wounds have distinct microbiomes from intact [96] Y. Lai, et al., High-coverage metabolomics uncovers microbiota-driven
skin, Microbiol Spectr. e0330022 (2022), https://doi.org/10.1128/ biochemical landscape of interorgan transport and gut-brain communication in
spectrum.03300-22. mice, Nat. Commun. 12 (2021) 6000.
[64] J.P. Hugot, et al., Association of NOD2 leucine-rich repeat variants with [97] W.S. Garrett, Cancer and the microbiota, Science 348 (2015) 80–86.
susceptibility to Crohn’s disease, Nature 411 (2001) 599–603. [98] S.J.D. O’Keefe, Diet, microorganisms and their metabolites, and colon cancer,
[65] Y. Ogura, et al., A frameshift mutation in NOD2 associated with susceptibility to Nat. Rev. Gastroenterol. Hepatol. 13 (2016) 691–706.
Crohn’s disease, Nature 411 (2001) 603–606. [99] P. Liu, et al., The role of short-chain fatty acids in intestinal barrier function,
[66] E. Fleming, et al., Cultivation of common bacterial species and strains from inflammation, oxidative stress, and colonic carcinogenesis, Pharm. Res 165
human skin, oral, and gut microbiota, BMC Microbiol 21 (2021) 278. (2021), 105420.
[67] X. He, et al., Cultivation of a human-associated TM7 phylotype reveals a reduced [100] K.M. Maslowski, et al., Regulation of inflammatory responses by gut microbiota
genome and epibiotic parasitic lifestyle, Proc. Natl. Acad. Sci. USA 112 (2015) and chemoattractant receptor GPR43, Nature 461 (2009) 1282–1286.
244–249. [101] N. Arpaia, et al., Metabolites produced by commensal bacteria promote
[68] B. Bor, et al., Insights obtained by culturing saccharibacteria with their bacterial peripheral regulatory T-cell generation, Nature 504 (2013) 451–455.
hosts, J. Dent. Res 99 (2020) 685–694. [102] Y. Furusawa, et al., Commensal microbe-derived butyrate induces the
[69] K. Nagashima, et al., Mapping the T cell repertoire to a complex gut bacterial differentiation of colonic regulatory T cells, Nature 504 (2013) 446–450.
community, 2022.05.04, bioRxiv (2022), 490632, https://doi.org/10.1101/ [103] P.V. Chang, L. Hao, S. Offermanns, R. Medzhitov, The microbial metabolite
2022.05.04.490632. butyrate regulates intestinal macrophage function via histone deacetylase
[70] E. van Nood, et al., Duodenal infusion of donor feces for recurrent Clostridium inhibition, Proc. Natl. Acad. Sci. USA 111 (2014) 2247–2252.
difficile, N. Engl. J. Med 368 (2013) 407–415. [104] K.Y.C. Fung, L. Cosgrove, T. Lockett, R. Head, D.L. Topping, A review of the
[71] M. Dsouza, et al., Colonization of the live biotherapeutic product VE303 and potential mechanisms for the lowering of colorectal oncogenesis by butyrate, Br.
modulation of the microbiota and metabolites in healthy volunteers, Cell Host J. Nutr. 108 (2012) 820–831.
Microbe 30 (583–598) (2022), e8. [105] Y. Cheng, Z. Ling, L. Li, The Intestinal Microbiota and Colorectal Cancer, Front
[72] E.O. Petrof, et al., Stool substitute transplant therapy for the eradication of Immunol. 11 (2020), 615056.
Clostridium difficile infection: ‘RePOOPulating’ the gut, Microbiome 1 (2013) 3. [106] Y. Tang, Y. Chen, H. Jiang, G.T. Robbins, D. Nie, G-protein-coupled receptor for
[73] D. Kao, et al., The effect of a microbial ecosystem therapeutic (MET-2) on short-chain fatty acids suppresses colon cancer, Int J. Cancer 128 (2011)
recurrent Clostridioides difficile infection: a phase 1, open-label, single-group 847–856.
trial, Lancet Gastroenterol. Hepatol. 6 (2021) 282–291. [107] A.J. Brown, et al., The Orphan G protein-coupled receptors GPR41 and GPR43 are
[74] F. Cold, C.K. Svensson, A.M. Petersen, L.H. Hansen, M. Helms, Long-Term Safety activated by propionate and other short chain carboxylic acids, J. Biol. Chem. 278
Following Faecal Microbiota Transplantation as a Treatment for Recurrent (2003) 11312–11319.
Clostridioides difficile Infection Compared with Patients Treated with a Fixed [108] V. Ganapathy, M. Thangaraju, P.D. Prasad, P.M. Martin, N. Singh, Transporters
Bacterial Mixture: Results from a Retrospective Cohort Study, Cells 11 (2022). and receptors for short-chain fatty acids as the molecular link between colonic
bacteria and the host, Curr. Opin. Pharm. 13 (2013) 869–874.

9
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

[109] N. Singh, et al., Activation of Gpr109a, receptor for niacin and the commensal [146] M.A. Koch, et al., Maternal IgG and IgA Antibodies Dampen Mucosal T Helper,
metabolite butyrate, suppresses colonic inflammation and carcinogenesis, Cell Responses Early Life. Cell 165 (2016) 827–841.
Immunity 40 (2014) 128–139. [147] R. Ahmad, M.F. Sorrell, S.K. Batra, P. Dhawan, A.B. Singh, Gut permeability and
[110] L.B. Bindels, et al., Gut microbiota-derived propionate reduces cancer cell mucosal inflammation: bad, good or context dependent, Mucosal Immunol. 10
proliferation in the liver, Br. J. Cancer 107 (2012) 1337–1344. (2017) 307–317.
[111] A. Belcheva, et al., Gut microbial metabolism drives transformation of MSH2- [148] L.R. Lopez, J.-H. Ahn, T. Alves, J.C. Arthur, Microenvironmental Factors that
deficient colon epithelial cells, Cell 158 (2014) 288–299. Shape Bacterial Metabolites in Inflammatory Bowel Disease, Front Cell Infect.
[112] R.E. Ley, P.J. Turnbaugh, S. Klein, J.I. Gordon, Microbial ecology: human gut Microbiol 12 (2022), 934619.
microbes associated with obesity, Nature 444 (2006) 1022–1023. [149] L. Robrahn, L. Jiao, T. Cramer, Barrier integrity and chronic inflammation
[113] T.M. Loo, et al., Gut Microbiota Promotes Obesity-Associated Liver Cancer mediated by HIF-1 impact on intestinal tumorigenesis, Cancer Lett. 490 (2020)
through PGE(2)-Mediated Suppression of Antitumor Immunity, Cancer Disco 7 186–192.
(2017) 522–538. [150] A. Piechota-Polanczyk, J. Fichna, Review article: the role of oxidative stress in
[114] P. Marzullo, et al., Spot-light on microbiota in obesity and cancer, Int J. Obes. pathogenesis and treatment of inflammatory bowel diseases, Naunyn Schmiede
(Lond. ) 45 (2021) 2291–2299. Arch. Pharm. 387 (2014) 605–620.
[115] J. Park, T.S. Morley, M. Kim, D.J. Clegg, P.E. Scherer, Obesity and cancer– [151] M.T. Abreu, R.M.J. Peek, Gastrointestinal malignancy and the microbiome,
mechanisms underlying tumour progression and recurrence, Nat. Rev. Gastroenterology 146 (1534–1546) (2014), e3.
Endocrinol. 10 (2014) 455–465. [152] R.F. Schwabe, C. Jobin, The microbiome and cancer, Nat. Rev. Cancer 13 (2013)
[116] R.W. O’Rourke, Obesity and cancer: at the crossroads of cellular metabolism and 800–812.
proliferation, Surg. Obes. Relat. Dis. 10 (2014) 1208–1219. [153] J.A. DiDonato, F. Mercurio, M. Karin, NF-κB and the link between inflammation
[117] G. Brandi, et al., Microbiota, NASH, HCC and the potential role of probiotics, and cancer, Immunol. Rev. 246 (2012) 379–400.
Carcinogenesis 38 (2017) 231–240. [154] G. Zhang, R. Chen, J.D. Rudney, Streptococcus cristatus modulates the
[118] M.F. Gregor, G.S. Hotamisligil, Inflammatory mechanisms in obesity, Annu Rev. Fusobacterium nucleatum-induced epithelial interleukin-8 response through the
Immunol. 29 (2011) 415–445. nuclear factor-kappa B pathway, J. Periodontal Res 46 (2011) 558–567.
[119] R. Mirzaei, et al., Role of microbiota-derived short-chain fatty acids in cancer [155] M.R. Milward, et al., Differential activation of NF-kappaB and gene expression in
development and prevention, Biomed. Pharm. 139 (2021), 111619. oral epithelial cells by periodontal pathogens, Clin. Exp. Immunol. 148 (2007)
[120] S. Yoshimoto, et al., Obesity-induced gut microbial metabolite promotes liver 307–324.
cancer through senescence secretome, Nature 499 (2013) 97–101. [156] E. Allen-Vercoe, J. Strauss, K. Chadee, Fusobacterium nucleatum: an emerging gut
[121] V. Matson, C.S. Chervin, T.F. Gajewski, Cancer and the Microbiome-Influence of pathogen? Gut Microbes 2 (2011) 294–298.
the Commensal Microbiota on Cancer, Immune Responses, and Immunotherapy, [157] M.R. Rubinstein, et al., Fusobacterium nucleatum promotes colorectal
Gastroenterology 160 (2021) 600–613. carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA
[122] C. Ma, et al., Gut microbiome-mediated bile acid metabolism regulates liver adhesin, Cell Host Microbe 14 (2013) 195–206.
cancer via NKT cells, Science 360 (2018). [158] S.M.N. Udden, et al., NOD2 Suppresses Colorectal Tumorigenesis via
[123] S. Han, et al., A metabolomics pipeline for the mechanistic interrogation of the Downregulation of the TLR Pathways, Cell Rep. 19 (2017) 2756–2770.
gut microbiome, Nature 595 (2021) 415–420. [159] M.H. Zaki, et al., The NOD-like receptor NLRP12 attenuates colon inflammation
[124] M. Wang, et al., Strain dropouts reveal interactions that govern the metabolic and tumorigenesis, Cancer Cell 20 (2011) 649–660.
output of the gut microbiome, 2022.07.25, bioRxiv (2022), 501461, https://doi. [160] I.C. Allen, et al., The NLRP3 inflammasome functions as a negative regulator of
org/10.1101/2022.07.25.501461. tumorigenesis during colitis-associated cancer, J. Exp. Med 207 (2010)
[125] A.C. Tolonen, et al., Synthetic glycans control gut microbiome structure and 1045–1056.
mitigate colitis in mice, Nat. Commun. 13 (2022) 1244. [161] K.-M. Lin, et al., IRAK-1 bypasses priming and directly links TLRs to rapid NLRP3
[126] M.L. Patnode, et al., Strain-level functional variation in the human gut microbiota inflammasome activation, Proc. Natl. Acad. Sci. USA 111 (2014) 775–780.
based on bacterial binding to artificial food particles, Cell Host Microbe 29 [162] S.I. Grivennikov, et al., Adenoma-linked barrier defects and microbial products
(664–673) (2021), e5. drive IL-23/IL-17-mediated tumour growth, Nature 491 (2012) 254–258.
[127] N.A. Pudlo, et al., Phenotypic and Genomic Diversification in Complex [163] S. Wu, et al., A human colonic commensal promotes colon tumorigenesis via
Carbohydrate-Degrading Human Gut Bacteria, mSystems 7 (2022), e0094721. activation of T helper type 17 T cell responses, Nat. Med 15 (2009) 1016–1022.
[128] L.C.-H. Yu, Microbiota dysbiosis and barrier dysfunction in inflammatory bowel [164] E. Elinav, et al., Inflammation-induced cancer: crosstalk between tumours,
disease and colorectal cancers: exploring a common ground hypothesis, immune cells and microorganisms, Nat. Rev. Cancer 13 (2013) 759–771.
J. Biomed. Sci. 25 (2018) 79. [165] T. Irrazábal, A. Belcheva, S.E. Girardin, A. Martin, D.J. Philpott, The multifaceted
[129] M.A. Fischbach, Microbiome: focus on causation and mechanism, Cell 174 (2018) role of the intestinal microbiota in colon cancer, Mol. Cell 54 (2014) 309–320.
785–790. [166] A.E. Overacre-Delgoffe, et al., Microbiota-specific T follicular helper cells drive
[130] M.C. Andrews, et al., Gut microbiota signatures are associated with toxicity to tertiary lymphoid structures and anti-tumor immunity against colorectal cancer,
combined CTLA-4 and PD-1 blockade, Nat. Med 27 (2021) 1432–1441. Immunity 54 (2812–2824) (2021), e4.
[131] T.K. Pedersen, et al., The CD4(+) T cell response to a commensal-derived epitope [167] N. Li, S.I. Grivennikov, M. Karin, The unholy trinity: inflammation, cytokines, and
transitions from a tolerant to an inflammatory state in Crohn’s disease, Immunity STAT3 shape the cancer microenvironment, Cancer Cell 19 (2011) 429–431.
S1074–7613 (22) (2022) 00410–00411, https://doi.org/10.1016/j. [168] R. Niehus, N.M. Oliveira, A. Li, A.G. Fletcher, K.R. Foster, The evolution of
immuni.2022.08.016. strategy in bacterial warfare via the regulation of bacteriocins and antibiotics,
[132] M.E. Perez-Muñoz, P. Joglekar, Y.-J. Shen, K.Y. Chang, D.A. Peterson, Elife 10 (2021).
Identification and Phylogeny of the First T Cell Epitope Identified from a Human [169] L.-S. Ma, A. Hachani, J.-S. Lin, A. Filloux, E.-M. Lai, Agrobacterium tumefaciens
Gut Bacteroides Species, PLoS One 10 (2015), e0144382. deploys a superfamily of type VI secretion DNase effectors as weapons for
[133] Y. Yang, et al., Focused specificity of intestinal TH17 cells towards commensal interbacterial competition in planta, Cell Host Microbe 16 (2014) 94–104.
bacterial antigens, Nature 510 (2014) 152–156. [170] J.E. Silpe, J.W.H. Wong, S.V. Owen, M. Baym, E.P. Balskus, The bacterial toxin
[134] M. Xu, et al., c-MAF-dependent regulatory T cells mediate immunological colibactin triggers prophage induction, Nature 603 (2022) 315–320.
tolerance to a gut pathobiont, Nature 554 (2018) 373–377. [171] J.-P. Nougayrède, et al., Escherichia coli induces DNA double-strand breaks in
[135] E. Sefik, et al., MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce eukaryotic cells, Science 313 (2006) 848–851.
a distinct population of RORγ+ regulatory T cells, Science 349 (2015) 993–997. [172] J. Putze, et al., Genetic structure and distribution of the colibactin genomic island
[136] P.J.P.L. Teixeira, et al., Specific modulation of the root immune system by a among members of the family Enterobacteriaceae, Infect. Immun. 77 (2009)
community of commensal bacteria, Proc. Natl. Acad. Sci. USA 118 (2021). 4696–4703.
[137] Gardnerella vaginalis alters cervicovaginal epithelial cell function through [173] E. Buc, et al., High prevalence of mucosa-associated E. coli producing
microbe-specific immune responses. vol. 10, 2022. cyclomodulin and genotoxin in colon cancer, PLoS One 8 (2013), e56964.
[138] T. Rollenske, et al., Parallelism of intestinal secretory IgA shapes functional [174] M. Prorok-Hamon, et al., Colonic mucosa-associated diffusely adherent afaC+
microbial fitness, Nature 598 (2021) 657–661. Escherichia coli expressing lpfA and pks are increased in inflammatory bowel
[139] C. Yang, et al., Fecal IgA levels are determined by strain-level differences in disease and colon cancer, Gut 63 (2014) 761–770.
bacteroides ovatus and are modifiable by gut microbiota manipulation, Cell Host [175] J.C. Arthur, et al., Intestinal inflammation targets cancer-inducing activity of the
Microbe 27 (467–475) (2020), e6. microbiota, Science 338 (2012) 120–123.
[140] D.A. Peterson, N.P. McNulty, J.L. Guruge, J.I. Gordon, IgA response to symbiotic [176] J.C. Arthur, et al., Microbial genomic analysis reveals the essential role of
bacteria as a mediator of gut homeostasis, Cell Host Microbe 2 (2007) 328–339. inflammation in bacteria-induced colorectal cancer, Nat. Commun. 5 (2014)
[141] S. Hapfelmeier, et al., Reversible microbial colonization of germ-free mice reveals 4724.
the dynamics of IgA immune responses, Science 328 (2010) 1705–1709. [177] L. Guerra, R. Guidi, T. Frisan, Do bacterial genotoxins contribute to chronic
[142] N.W. Palm, et al., Immunoglobulin A coating identifies colitogenic bacteria in inflammation, genomic instability and tumor progression? FEBS J. 278 (2011)
inflammatory bowel disease, Cell 158 (2014) 1000–1010. 4577–4588.
[143] K.L. Alexander, et al., Human Microbiota Flagellins Drive Adaptive Immune [178] Z. He, et al., Campylobacter jejuni promotes colorectal tumorigenesis through the
Responses in Crohn’s Disease, Gastroenterology 161 (522–535) (2021), e6. action of cytolethal distending toxin, Gut 68 (2019) 289–300.
[144] J.J. Bunker, A. Bendelac, IgA Responses to Microbiota, Immunity 49 (2018) [179] H. Tsoi, et al., Peptostreptococcus anaerobius Induces Intracellular Cholesterol
211–224. Biosynthesis in Colon Cells to Induce Proliferation and Causes Dysplasia in Mice,
[145] M.Y. Zeng, et al., Gut Microbiota-Induced Immunoglobulin G Controls Systemic Gastroenterology 152 (1419–1433) (2017), e5.
Infection by Symbiotic Bacteria and Pathogens, Immunity 44 (2016) 647–658.

10
T. Plitt and J.J. Faith Seminars in Immunology 66 (2023) 101735

[180] X. Wang, et al., Enterococcus faecalis induces aneuploidy and tetraploidy in [195] A.D. Kostic, et al., Genomic analysis identifies association of Fusobacterium with
colonic epithelial cells through a bystander effect, Cancer Res 68 (2008) colorectal carcinoma, Genome Res 22 (2012) 292–298.
9909–9917. [196] M. Castellarin, et al., Fusobacterium nucleatum infection is prevalent in human
[181] C.M. Dejea, et al., Patients with familial adenomatous polyposis harbor colonic colorectal carcinoma, Genome Res 22 (2012) 299–306.
biofilms containing tumorigenic bacteria, Science 359 (2018) 592–597. [197] C.L. Enterotoxigenic Sears, Bacteroides fragilis: a rogue among symbiotes, Clin.
[182] L. Chung, et al., Bacteroides fragilis toxin coordinates a pro-carcinogenic Microbiol Rev. 22, 349–369, Table Contents (2009).
inflammatory cascade via targeting of colonic epithelial cells, Cell Host Microbe [198] S. Wu, K.-J. Rhee, M. Zhang, A. Franco, C.L. Sears, Bacteroides fragilis toxin
23 (203–214) (2018), e5. stimulates intestinal epithelial cell shedding and gamma-secretase-dependent E-
[183] B.C. Nelson, et al., Emerging metrology for high-throughput nanomaterial cadherin cleavage, J. Cell Sci. 120 (2007) 1944–1952.
genotoxicology, Mutagenesis 32 (2017) 215–232. [199] A. Boleij, et al., The Bacteroides fragilis toxin gene is prevalent in the colon
[184] S. Dubbert, B. Klinkert, M. Schimiczek, T.M. Wassenaar, R. Bünau, von. No mucosa of colorectal cancer patients, Clin. Infect. Dis. 60 (2015) 208–215.
Genotoxicity Is Detectable for Escherichia coli Strain Nissle 1917 by Standard In [200] S. Wu, P.J. Morin, D. Maouyo, C.L. Sears, Bacteroides fragilis enterotoxin induces
Vitro and In Vivo Tests. Eur, J. Microbiol Immunol. (Bp) 10 (2020) 11–19. c-Myc expression and cellular proliferation, Gastroenterology 124 (2003)
[185] J.J. Mousa, et al., MATE transport of the E. coli-derived genotoxin colibactin, Nat. 392–400.
Microbiol 1 (2016) 15009. [201] U. Dutta, P.K. Garg, R. Kumar, R.K. Tandon, Typhoid carriers among patients with
[186] T. Irrazabal, et al., Limiting oxidative DNA damage reduces microbe-induced gallstones are at increased risk for carcinoma of the gallbladder, Am. J.
colitis-associated colorectal cancer, Nat. Commun. 11 (2020) 1802. Gastroenterol. 95 (2000) 784–787.
[187] V. Graillot, et al., Genotoxicity of Cytolethal Distending Toxin (CDT) on Isogenic [202] E.C. Lazcano-Ponce, et al., Epidemiology and molecular pathology of gallbladder
Human Colorectal Cell Lines: Potential Promoting Effects for Colorectal cancer, CA Cancer J. Clin. 51 (2001) 349–364.
Carcinogenesis, Front Cell Infect. Microbiol 6 (2016) 34. [203] F. de la Gala Sánchez, J.J. Jorge Gómez, P. García Méndez, M. González Estecha,
[188] Cao, Y. et al. Commensal microbiota from patients with inflammatory bowel [Ferritin and iron deficiency anemias], Med Interna 6 (1989) 133–136.
disease produce genotoxic metabolites. Science 378, eabm3233 (2022). [204] R. Lu, et al., Presence of Salmonella AvrA in colorectal tumor and its precursor
[189] O. Silva-García, J.J. Valdez-Alarcón, V.M. Baizabal-Aguirre, Wnt/β-Catenin lesions in mouse intestine and human specimens, Oncotarget 8 (2017)
Signaling as a Molecular Target by Pathogenic Bacteria, Front Immunol. 10 55104–55115.
(2019) 2135. [205] J.H. Sellin, S. Umar, J. Xiao, A.P. Morris, Increased beta-catenin expression and
[190] M.R. Rogan, L.L. Patterson, J.Y. Wang, J.W. McBride, Bacterial Manipulation of nuclear translocation accompany cellular hyperproliferation in vivo, Cancer Res
Wnt Signaling: A Host-Pathogen Tug-of-Wnt, Front Immunol. 10 (2019) 2390. 61 (2001) 2899–2906.
[191] S. Alzahrani, et al., Effect of Helicobacter pylori on gastric epithelial cells, World [206] J.V. Newman, T. Kosaka, B.J. Sheppard, J.G. Fox, D.B. Schauer, Bacterial
J. Gastroenterol. 20 (2014) 12767–12780. infection promotes colon tumorigenesis in Apc(Min/+) mice, J. Infect. Dis. 184
[192] M. Hatakeyama, Structure and function of Helicobacter pylori CagA, the first- (2001) 227–230.
identified bacterial protein involved in human cancer, Proc. Jpn Acad. Ser. B [207] B. Udayasuryan, et al., Fusobacterium nucleatum induces proliferation and
Phys. Biol. Sci. 93 (2017) 196–219. migration in pancreatic cancer cells through host autocrine and paracrine
[193] L.F. Jiménez-Soto, R. Haas, The CagA toxin of Helicobacter pylori: abundant signaling, Sci. Signal 15 (2022).
production but relatively low amount translocated, Sci. Rep. 6 (2016) 23227. [208] G.F. Gao, et al., Before and After: Comparison of Legacy and Harmonized TCGA
[194] A.D. Kostic, et al., Fusobacterium nucleatum potentiates intestinal tumorigenesis Genomic Data Commons’ Data, Cell Syst. 9 (24–34) (2019), e10.
and modulates the tumor-immune microenvironment, Cell Host Microbe 14 [209] ImmGen at 15. Nat Immunol 21, 700–703, 2020.
(2013) 207–215.

11

You might also like