You are on page 1of 21

Review

Good Gone Bad: One Toxin Away


From Disease for Bacteroides fragilis

Ezequiel Valguarnera and Juliane Bubeck Wardenburg


Department of Pediatrics, Washington University School of Medicine, 660 S. Euclid Ave. Box 8208, St. Louis, MO 63110

Correspondence to Juliane Bubeck Wardenburg: jbubeck@wustl.edu


https://doi.org/10.1016/j.jmb.2019.12.003
Edited by John Ralston Brannon

Abstract
The human gut is colonized by hundreds of trillions of microorganisms whose acquisition begins during early
infancy. Species from the Bacteroides genus are ubiquitous commensals, comprising about thirty percent of
the human gut microbiota. Bacteroides fragilis is one of the least abundant Bacteroides species, yet is the
most common anaerobe isolated from extraintestinal infections in humans. A subset of B. fragilis strains carry
a genetic element that encodes a metalloprotease enterotoxin named Bacteroides fragilis toxin, or BFT.
Toxin-bearing strains, or Enterotoxigenic B. fragilis (ETBF) cause acute and chronic intestinal disease in
children and adults. Despite this association with disease, around twenty percent of the human population
appear to be asymptomatic carriers of ETBF. BFT damages the colonic epithelial barrier by inducing cleavage
of the zonula adherens protein E-cadherin and initiating a cell signaling response characterized by
inflammation and c-Myc-dependent pro-oncogenic hyperproliferation. As a consequence, mice harboring
genetic mutations that predispose to colonic inflammation or tumor formation are uniquely susceptible to toxin-
mediated injury. The recent observation of ETBF-bearing biofilms in colon biopsies from humans with colon
cancer susceptibility loci strongly suggests that ETBF is a driver of colorectal cancer. This article will address
ETBF biology from a host-pathobiont perspective, including clinical data, analysis of molecular mechanisms of
disease, and the complex ecological context of the human gut.
© 2019 Published by Elsevier Ltd.

Human beings are persistently colonized by commensal during the lifetime of an individual, with
commensal microorganisms. The gut microbiota, in colonization occurring during early childhood
particular, is composed of tens of trillions of micro- [12e14]. Nevertheless, a subset of strains can
organisms and is normally acquired from birth to produce a proteolytic enterotoxin, named B.fragilis
around 3 years of age [1]. Bacteroides fragilis is a toxin (BFT), or fragilysin, that causes secretory
ubiquitous member of the human gut microbiota. It diarrhea and colonic epithelial damage [15]. BFT is
belongs to the phylum Bacteroidetes, which together among the most studied virulence factors of
with the phylum Firmicutes constitutes about 80% of B. fragilis, and current evidence suggests that this
the total human gut microbiota [2,3]. B. fragilis is a toxin may be a driver for chronic colitis and colorectal
commensal organism that can become an opportu- cancer [16e18]. Toxin-producing strains, or enter-
nistic pathogen in certain individuals. Although otoxigenic B. fragilis (ETBF), were discovered in the
B. fragilis comprises around 0.1e0.5% of total gut 1980s and were found to be associated with diarrhea
bacteria, it is the most frequently isolated anaerobe in lambs, calves, pigs, foals, and humans and also
from peritoneal and abdominal abscesses, as well found to be present in sewage waters [19e24]. Since
as from samples of bloodstream infections [4]. This those initial reports, multiple studies have enhanced
species became notorious in the 1970s because our understanding of ETBF as a relevant pathogen in
reports have shown its relevance in peritoneal, humans and BFT as the key virulence factor in
abdominal, intestinal, and blood infections, as well disease. Humans are variably colonized with non-
as its propensity to develop antibiotic resistance toxigenic B. fragilis (NTBF) or ETBF strains, and
[5e11]. Most B. fragilis strains will remain as a most frequently, a single B. fragilis strain type is

0022-2836/© 2019 Published by Elsevier Ltd. Journal of Molecular Biology (2020) 432, 765e785
766 Good gone bad

found in fecal samples from individual healthy the following question arisesdwhat is the benefit of
donors [14]. Presently, knowledge of genetic deter- toxin production by B. fragilis in an organism that
minants of human colonization by B. fragilis is in its exhibits a commensal existence? Several plausible
infancy. The host and microbial factors that govern answers to this question can be put forth: (1)
temporal and spatial plasticity of the B. fragilis niche intoxication facilitates colonic niche acquisition and
are not understood; similarly, determinants respon- survival. A report from our group showed that a
sible for progression from asymptomatic carriage of particular strain of ETBF (43859) can colonize a
B. fragilis toward disease remain undefined. In this niche previously occupied by NTBF (TM4000) in a
work, we will review current knowledge on toxigenic toxin-dependent manner [45]. The presence of the
B. fragilis (ETBF) and discuss the dual nature of this toxin was not, however, a universal determinant of
organism as both a commensal and pathogen in the intraluminal niche acquisition or competition, which
context of a complex ecological scenario. also relies in part on the B. fragilis type VI secretion
system (T6SS) and other genetic determinants
[46e54]. A precedent for toxin-mediated modulation
Colonization of Humans by ETBF of the bacterial niche to facilitate survival has
recently been demonstrated for Vibriocholerae [55].
ETBF has been identified in stool samples from (2) Intoxication facilitates transmission of ETBF. As
healthy individuals and patients with diarrhea. previously mentioned, in children aged between 1
Interestingly, the prevalence of ETBF is about and 5 years, the ETBF count is increased in patients
twice as high in patients (20e30%) than in healthy with diarrhea. Toxin production and the associated
individuals (10e20%) [25e27]. Reports from distinct diarrhea may represent a key strategy for transmis-
urban and underdeveloped geographic locations sion of the pathogen from human to human, by
show higher percentages of ETBF colonization in increasing fecal-oral contamination. Human intest-
children older than 1 year with diarrhea than in age- inal pathogens such as V.cholerae and Salmonella
matched controls [25,28e32]. Interestingly, in chil- enterica use diarrhea as a means to transmit
dren younger than 1 year, prevalence is the lowest in infection between human hosts, so this could also
the general population and is not associated with be the case for ETBF [56,57]. (3) Intoxication permits
diarrhea, suggesting that the developmental trajec- an extraintestinal lifestyle for B. fragilis. As a leading
tory is important for diarrheal association to occur cause of anaerobic infection, B. fragilis exhibits the
[28,32]. An association of ETBF with chronic ability to invade multiple tissue sites within the body
intestinal disease has been established for more [58]. Studies that have investigated the presence of
than 20 years, the first reported in patients with ETBF in extraintestinal infections have not reached a
inflammatory bowel disease (IBD) [33]. ETBF is also clear consensus; however, they suggest that ETBF
positively associated with ulcerative colitis and may be more represented in bloodstream isolates
colonic neoplasia [34e41]. The association of and samples of vaginal infections than in other
ETBF in patients with colorectal cancer (CRC) isolation sites [59e65]. BFT has been shown to be
includes sporadic and familial cases, indicating that toxic in vitro to kidney and lung epithelial cells as well
progression of this disease, regardless of the onset, as to the endothelium, suggesting a potential role in
has physiologic commonalities. The disparity extraintestinal infections [66,67]. Although other
between high asymptomatic carriage of ETBF in genetic determinants of colonization, including cap-
human populations and the low number of cases of sular polysaccharide, have been implicated in
ETBF-associated IBD and sporadic CRC suggests extraintestinal infections and abscess formation
that pathogenicity is not stochastic and depends on [68], such roles have not been clearly defined for
unknown host susceptibility determinants. It is BFT. A considerable amount of epidemiologic
currently not known if ETBF strains typically produce investigation focused within the human population
enterotoxin in asymptomatic carriers [42]. Studies and paired mechanistic analyses in model systems
measuring amounts of secreted BFT in both healthy will be needed to provide clarity on the potential
and diseased individuals will be required to establish evolutionary advantage of maintenance of BFT and
a functional association between carriage of ETBF its associated pathogenicity island within the
and specific manifestations of disease. Furthermore, B. fragilis genome.
the fact that NTBF accounts for the vast majority of
B. fragiliseassociated infections increases the com-
plexity of studies that will be required to define both Discovery of BFT and Toxin Activation
epidemiologic and mechanistic contributions of
ETBF to human disease. The first report of enterotoxicity by B. fragilis was
As non-toxigenic (NTBF readily colonizes the presented in 1984, describing diarrhea in lambs [19].
human colon and is suggested to benefit the At that time, there was a clear distinction between
development of the host T-cell response through enterotoxigenic and nonenterotoxigenic strains, but
studies conducted in murine model systems [43,44], the etiology of the diarrhea was unknown. In 1992,
Good gone bad 767

Weikel et al. [69] were the first to demonstrate that a


component in culture supernatants from enterotoxi-
genic strains was causing the secretory phenotype
previously observed when injecting lamb ileal loops
(LILs) with bacteria. The authors developed an in
vitro assay of BFT cell damage using colon
carcinoma HT29 cell lines, allowing for simultaneous
screening of multiple enterotoxigenic strains [69].
The toxin was initially purified from culture super-
natants as a 20-KDa protein and characterized as
enterotoxic and cytotoxic in LIL and HT29 models,
respectively. Partial cloning and expression of the
toxin revealed that it is a heat-labile metalloprotease
[70,71]. Diagnostics for detection of the toxin were
also developed, allowing for confirmation that the
enterotoxin is present in ETBF strains isolated from
Fig. 1. Known molecular mechanisms that govern
humans with ETBF-associated diarrhea [72,73]. The secretion of BFT. The RprX/RprY two-component system
complete sequence of the bft gene was cloned, and is activated under certain as-yet-unidentified conditions,
characterization of the amino acid sequence and with concomitant phosphorylation of RprY (RprY-P) and
biochemical analysis suggested that BFT is pro- binding to the bft promoter, repressing gene expression. In
duced as a protoxin that is processed releasing the a noneRprY-P situation, bft is expressed and translated
active C-terminal domain into the extracellular milieu as a signal peptide-containing protein into the periplasmic
[74,75]. Further studies showed that cleavage of the space. Pre-BFT is most likely a lipoprotein that gets
protoxin is not dependent on the zinc-binding motif of transported to the outer membrane. Surface-exposed pre-
the metalloprotease domain and that the whole C- BFT can be cleaved by Fragipain or host gut proteases,
releasing the active metalloprotease C-terminal domain
terminal domain is essential for toxin activity [76,77].
into the extracellular milieu. BFT, Bacteroides fragilis toxin;
Detailed structural analysis determined that the N- Fpn, fragipain.
terminal prodomain is likely involved in the secretion
of the protoxin through the cell envelope and in the
inhibition of toxin activity within the bacterial cell [78].
Interestingly, the N-terminal domain contains a fied, bft-1, bft-2, and bft-3 [62,82,83]. Most ETBF
lipoprotein signal peptide representing a unique strains carry a single bft variant; each variant
fold, whereas the C-terminal metalloprotease demonstrates different degrees of potency in vitro
domain is a xenolog of eukaryotic A Disintegrin and in germ-free mice. Among these, BFT-2 exhibits
and Metalloprotease (ADAM) proteases, suggesting the greatest potential to elicit tissue damage [84e86].
horizontal acquisition of bft by B. fragilis [78]. As Population-wide carriage has been studied, and BFT-
shown in Fig. 1, cleavage of the protoxin can occur in 1 was found to be the most widely distributed toxin
vivo by host proteases from the intestinal lumen. variant within ETBF human isolates, whereas BFT-3
Interestingly, in bloodstream, BFT can only be seems to be geographically restricted to southeast
activated by a single endogenous bacterial cysteine Asia [31,62,87,88].
protease named fragipain (Fpn) [67,79]. An ETBF The BfPAI contains 12-bp repeats on both ends,
fpn mutant strain was unable to cause lethality in and it is always inserted in the same genomic
mice when bacteria were administered intrave- localization. A 17-bp GC-rich sequence is also found
nously, showing the importance of Fpn in the context at the putative insertion site in NTBF strains [80]. The
of ETBF-mediated sepsis [67]. The fpn gene is BfPAI is present within the conjugative transposon
present in most NTBF and ETBF strains, suggesting CTn86, supporting the hypothesis of horizontal
an additional role of Fpn beyond BFT activation. acquisition of the BfPAI by ETBF strains [89e92].
Genome sequencing and comparison indicates high
genetic diversity between ETBF strains [92,93].
Genomic Context and Transcriptional Some ETBF strains are more closely related to
Regulation NTBF strains than to other ETBF strains, further
supporting that the BfPAI has been acquired through
Studies on the genetic locus that encodes BFT have multiple independent horizontal transfer events [92].
been crucial to understand its origin and distribution Thus, the only known genetic determinant that
within strains. The bft gene is part of the ~6-kbp allows for differentiation between ETBF and NTBF
B. fragilis pathogenicity island (BfPAI). The BfPAI is strains is the presence or absence of the BfPAI,
absent in NTBF strains, and it also encodes another respectively.
metalloprotease (mpII), whose role in pathogenicity is The BfPAI contains a promoter sequence
unclear [80,81]. Three bft variants have been identi- upstream of the bft gene that controls transcription
768 Good gone bad

[94]. A key mechanism for control of toxin production


is regulation of expression by the two-component
system (TCS) RprX/RprY. The TCS is a common
strategy to couple environmental stimuli with gene
regulation in bacteria, in which a sensor histidine
kinase (RprX) detects specific stimuli and phosphor-
ylates a response regulator (RprY) that binds to
promoter regions in the bacterial genome inducing or
repressing gene expression [95]. Phosphorylated
RprY binds to the promoter upstream of the bft gene
and represses expression (Fig. 1). Overexpression
of RprY abrogates toxin expression, and deletion of
rprY causes increased nonregulated expression of
bft [96]. Hence, ETBF strains could be sensing
environmental cues to determine the optimal situa-
tion for toxin expression. BFT can be repressed by
fermentable sugars and induced by heat and
oxidative stress [45]. Although RprX/Y seems the
key to bft regulation in vivo, stimuli for this TCS
inside the host remain unknown.

Toxin-Host Interactions Fig. 2. BFT-host cell interactions. BFT binds to colonic


epithelial cells (CECs) through an unknown receptor and
Since the discovery of BFT, the molecular triggers cleavage of E-cadherin. BFT, Bacteroides fragilis
mechanisms that govern damage of host cells toxin; ETBF, enterotoxigenic Bacteroides fragilis.
have been described. When the HT29 cell model
was established, it was observed that BFT caused promotes migration of beta-catenin to the nucleus
cell rounding and shedding, presumed to result from [105]. In addition, E-cadherin cleavage by BFT
damage to the intercellular junction [66,71]. Morpho- triggers induction of mitogen-activated protein
logical changes in BFT-treated HT29 cells were kinases (MAPKs) and the NF-kappa B pathway,
found to be a product of F- and G-actin rearrange- thus increasing secretion of interleukin (IL)-8, a
ment [97,98]. BFT binds to an unidentified cell chemokine that attracts polymorphonuclear cells
receptor in a protease-dependent manner and [106e111]. NF-kappa B activation controls fluid
induces cleavage of the extracellular domain of the secretion of intestinal cells through induction of
zonula adherens protein E-cadherin; this cleavage COX2 and an increase in prostaglandin E2 levels
event occurs only in the context of intact cells (Fig. 2) [112]. COX2 and heme oxygenase-1 induction by
[99e101]. Given the similarity of BFT to ADAM10, it BFT is related to a delay of apoptosis in intestinal
is possible that BFT contributes directly to E- epithelial cells [113,114]. BFT can also induce
cadherin cleavage [78]. As there is no evidence for mechanisms of host defense such as beta-defensin
direct processing of cellular E-cadherin by BFT yet, 2 and expression of the siderophore-binding anti-
an alternative hypothesis is that activity of BFT microbial protein lipocalin-2 [115,116] and also
toward its receptor or another host protein could increases autophagy in human umbilical vein
trigger a signaling pathway responsible for the loss endothelial cells through the MAPK, AP-1 [117].
of E-cadherin (Fig. 2) [100]. A study using human Signaling pathways affected by the toxin cause
colonic biopsies incubated with BFT on the luminal differential gene expression and epigenetic changes
or serosal side of the epithelium showed more in HT29 cells [118]. Variations in the host cell
damage on the latter, suggesting BFT receptor transcription profile and epigenetic marks are lost
polarization toward the basal cell region [102]. when toxin stimulation is withdrawn, suggesting that
BFT-dependent cleavage of E-cadherin causes continuous toxin secretion by ETBF could be a
loss of cell-cell contacts and cell rounding, which component of disease progression [118].
requires the intramembrane protease presenilin-1/ The relevance of mechanistic findings in cell
gamma secretase complex [103]. The intracellular culture systems has been elaborated in animal
domain of E-cadherin is normally bound to a- and b- models of ETBF-associated disease. Rabizadeh
catenins [104]. When b-catenin is dissociated from et al [119] and Rhee et al [120] showed for the first
E-cadherin, it can function as a transcription factor in time in specific pathogenefree (SPF) C57BL/6 mice
a Wnt-dependent manner, inducing cell proliferation that ETBF causes acute and persistent colitis
through activation of the c-Myc pathway [104] in mice, driven by cleavage of E-cadherin by BFT
(Fig. 3). BFT-mediated cleavage of E-cadherin in vivo. Activation of STAT3 in vivo can be observed
Good gone bad 769

main aspects of colonization: (1) the relevance of the


B. fragilis capsule and other factors in host-patho-
biont relationships and (2) mechanisms that mediate
interbacterial competition in the gut, in particular
T6SSs and secreted antimicrobial toxins.

Capsular Polysaccharide

The B. fragilis capsule was described in the 1970s


in the studies by Kasper [126], Kasper et al [127,129]
and Lindberg et al [128] as a distinctive feature of this
organism relative to other species from the genus
Bacteroides. These observations, coupled with the
fact that B. fragilis is the most commonly isolated
Bacteroides species from anaerobic infections in
humans despite its low relative abundance in the
microbiome, led to the hypothesis that the B. fragilis
capsule is linked to disease pathogenesis. Indeed,
studies have indicated that capsule reduces phago-
cytosis by immune cells, thereby increasing bacterial
fitness outside the colonic lumen [130]. Consistent
with this finding, capsular polysaccharide is the main
Fig. 3. Pro-oncogenic signaling by BFT. CECs contributor to extraintestinal abscess formation by
induce cMyc through b-catenin and STAT3 activating a B. fragilis [128,130e132].
cell proliferation program. cMyc also induces spermine B. fragilis harbors 8 distinct genomic loci, each of
oxidase (SMOX), in a BFT-dependent manner, generating
a source of ROS (Reactive Oxygen Species) that can
which encodes the required enzymes for synthesis
contribute to DNA damage. BFT, Bacteroides fragilis toxin; of a specific capsular polysaccharide variant.
CEC, colonic epithelial cell. Expression of these loci is regulated through
promoter inversion to “on” or “off” configurations
[133,134]. Capsular polysaccharide levels are
at 24 h after infection in mucosal immune cells and increased when B. fragilis is animal passaged,
colonic epithelial cells [121], inducing c-myc expres- indicating its key role in survival within the host
sion and concomitant cell proliferation (Fig. 3) [130]. Monocolonization experiments in germ-free
[122,123]. Colonic histology of acute colitis (2 days mice show that any capsular polysaccharide is
after infection) in SPF mice shows rupture of cell-cell sufficient for niche establishment [135]; however,
adhesions and epithelial exfoliation, with the pre- this finding does not indicate that all capsule variants
sence of immune cell infiltrates [121]. Conversely, are immunologically equivalent within the host or that
chronic colitis, observed between 7 days and up to a single capsule variant is sufficient for colonization
16 months after infection, shows progressive hyper- in a complex ecosystem [135,136]. B. fragilis cap-
plasia of the colonic crypts, consequent with BFT- sular polysaccharides are composed of repeating
dependent induction of a cell hyperproliferation units of zwitterionic glycans [132]. Polysaccharide A
program [120,121]. (PSA), unlike most glycan antigens, can be pre-
sented within the context of MHCII antigen-present-
ing cells, inducing formation of regulatory T cells that
Bacterial Factors in Colonization and contribute to immune tolerance toward B. fragilis
Disease [137e139]. PSA induces IL-10 production through
the Toll-like receptor (TLR) 2 pathway and represses
Species from the Bacteroides genus are acquired production of IL-17 in germ-free mice, promoting a
early in life and are commonly found in the lower low inflammation environment [44]. The lack of PSA
gastrointestinal tract [1]. There are many potential however does not decrease IL-10 in SPF mice
determinants of success for a given strain to become colonized with NTBF [140,141]. For ETBF, recent
established in its niche, including host diet, devel- research using snap-frozen samples from human
opment, antibiotic usage, and interactions with other colonic tissue biopsies showed a negative associa-
members of the microbiota [12,124,125]. Although tion between PSA expression and carriage of bft
external factors, such as the aforementioned ones, gene [140], suggesting the potential to exacerbate
are pivotal for colonization, B. fragilis carry genetic inflammation. Together, these studies suggest there
determinants of colonization that contribute to niche are likely strain- and context-dependent regulatory
occupancy by interacting with the host and microbial elements that modulate the host response to
competitors. In this section, we will emphasize two B. fragilis polysaccharides.
770 Good gone bad

Polysaccharide Utilization Loci Neuraminidase


A genetic screening by Lee et al [142] identified the Bacterial glycosidases are ubiquitous and are
“commensal colonization factor” (ccf) operon that is used by many organisms to degrade complex
conserved among many Bacteroides species and polysaccharides for nutritional and ecological pur-
confers stable niche acquisition in mice. The poses [154]. B. fragilis and other Bacteroides sp.
structure of the ccf operon resembles typical Can cleave sialic acid from host glycoproteins by
polysaccharide utilization loci (PULs) from Bacter- secreting neuraminidase [155,156]. Sialic acid is
oides [142]. PULs are specialized loci dedicated to commonly present on host glycoproteins [157]. The
the breakdown and assimilation of complex glycans; most studied neuraminidase from B. fragilis is
these constitute close to 20% of the genomic content encoded by the nanH gene. Deletion of nanH
of some Bacteroides species [143]. Different PULs renders a mutant strain that is outgrown by the
can process distinct polysaccharides, allowing Bac- wild-type strain in vitro and in vivo [158]. Sialic acid
teroides species to process both dietary and host release from host glycoproteins and utilization is
glycans [143e147]. Versatility in nutrient utilization likely to function as an extra measure of nutrient
increases the chances of survival during host diet versatility, crucial to a gut commensal. Neuramini-
changes; thus, PUL diversity within Bacteroides dase seems to also increase binding of B. fragilis to
species is the key to niche acquisition and stable mammalian epithelial cells, by releasing sialic acid
niche occupancy. Similar to most PULs, the ccf and uncovering other glycan moieties [159,160]. The
operon consists of a transcriptional regulator sigma/ presence of the nanH gene has also been used as a
antisigma factor pair (ccfA and ccfB) that controls means of additional taxonomic information for
gene expression. Downstream of ccfA/B is the classification of B. fragilis isolates from infection
TonB-dependent receptor that transports breakdown sites and stool samples [161].
products into the cell periplasm (ccfC) and the SusD-
like accessory lipoprotein (ccfD) that is required for
Proteases
control of nutrient transport by ccfC. The last gene in
the locus is a putative chitobiase (ccfE) that cleaves B. fragilis has been shown to produce other
a complex substrate polymer into oligomers for cell proteases than BFT and fragipain. A subset of
acquisition. Functional characterization of ccf cysteine proteases of the C10 family (BFP) was
showed that ccfA, ccfC, and ccfD are required for found in genomes of B. fragilis strains [162]. Genes
stable colonization, precluding secondary coloniza- bfp1e4 are present within mobile genetic elements,
tion events by Bacteroides from the same species. which is indicative of genetic acquisition by B. fragilis
On the contrary, ccfE is not required for ccf function, through horizontal transmission. bfp gene expres-
most likely owing to enzymatic redundancy from sion, particularly bfp4, is induced in vitro with
other B. fragilis gene products [142]. A recent study increased oxygen concentrations, suggesting a
showed that the ccf locus controls capsule variants, putative role of BFP in adaptation to environmental
repressing PSA and inducing polysaccharide C changes [163]. Bfp genes have been found both in
(PSC) expression. ccf function allows IgA-depen- bacterial isolates from infected sites and in stool
dent niche establishment in response to PSC, samples from healthy donors [162]; however, iso-
enabling B. fragilis cells to localize closer to the gut genic bfp deletion strains have not been evaluated to
epithelium [142,148]. The specific environmental assess the role of BFP in pathogenicity. A fibrinogen-
cues that induce ccf expression in vivo remain to degrading protease has also been identified in
be defined. B. fragilis, although its role in virulence is unknown
[164,165].
B. fragilis Hemolysins
Many gram-negative and gram-positive bacteria Adhesive Molecules
secrete enzymes that lyse red blood cells, known as Bacterial cells adhere to substrates via adhesins,
hemolysins [149e151]. B. fragilis is not an proteins that bind specifically to receptors from host
exception as many strains carry hemolysin ortholog cells [166,167]. Many adhesins are expressed as a
genes [152]. Hemolysins A and B (HlyA and HlyB, part of fimbriae or pili [168]. In commensal gut
respectively) have been shown to be enzymatically bacteria such as Bacteroides, binding to intestinal
active in vitro against red blood cells [152]. B. fragilis mucus and epithelial cells favors stable gut niche
mutant strains lacking genes hlyA/B show reduced colonization. In humans, piliated strains are more
fitness in vitro and in vivo, indicating that hemolysins commonly associated with abscess and healthy
may be involved in colonization [153]. At present, stool samples, whereas nonpiliated strains are
there is no clear evidence showing a specific role of enriched in isolates from blood infections [169].
hemolysins in the pathogenesis of disease caused Adhesive-piliated Bacteroides are more easily pha-
by B. fragilis. gocytosed by neutrophils; thus, loss of cell
Good gone bad 771

adhesiveness could promote extraintestinal disse- induction of many genes in the OSR pathway
mination and immune evasion [170]. Specific binding [185,186,198]. Another transcription factor bmoR
to red blood cells, or hemagglutination, has been has been reported as a component of OSR, inducing
observed more frequently in blood isolates than in genes required for the maintenance of the intracel-
abscess or healthy stool isolates [171e173]. Sialic lular redox state [199,200].
acid has been shown to be required for hemagglu-
tinin phenotypes, and it is proposed as the receptor
Response to Other Environmental Stimuli
for lectin-like adhesins from B. fragilis [159]. The
genetic basis for cell adhesiveness in B. fragilis still B. fragilis has shown to be responsive to the
remains unknown. presence of bile salts, increasing cell adhesion and
B. fragilis also presents extracellular matrix coaggregation in vitro [201,202]. There is no
(ECM)ebinding proteins, suggesting a role in extra- evidence of bile salt tolerance in Bacteroides as a
intestinal survival. Binding to fibronectin, the most pathogenic trait, rather than part of an environmental
abundant protein in the ECM, is mediated by a stress response. Transcriptional regulators such as
protein similar to a TonB-dependent receptor, the mar system mediate resistance to antimicrobials
BF1991 [174,175]. Surprisingly, a mutant strain and other environmental stressors [199,203].
lacking bf1991 is more adhesive to fibronectin that
the wild-type strain, indicative of redundant fibro-
nectin-binding proteins. Bf1991 mutants are more Lipopolysaccharide
susceptible in vitro to phagocytosis by macrophages In gram-negative bacteria, the outer layer of the
[175]. Binding of B. fragilis to laminin-1 and collagen- outer membrane is composed of phospholipids and
1 has also been reported [176e178]. Similar to other lipooligosaccharides or lipopolysaccharides (LOSs
human pathogens, B. fragilis can interact with or LPSs, respectively) [204]. LPS molecules are
plasma proteins related to coagulation [179]. BF- composed of a lipid moiety knows as Lipid A, a core
FBP is a 54-KDa protein that binds fibrinogen, the glycan, and an outer glycan [205,206]. Lipid A, or
major component of fibrin abscess formation [165]. endotoxin, triggers an inflammatory response,
Other proteins can bind plasminogen and high- dependent on recognition and signaling by TLR4 of
molecular-mass kininogen, yet the role in B. fragilis immune host cells [206e208]. The endotoxin of
manipulation of host coagulation is not clear Bacteroidesinduces a much lower degree of LPS/
[180,181]. The diversity of host targets that TLR4-mediated inflammation than Lipid A from
B. fragilis can potentially bind to allow us to species of Enterobacteria owing to differences in
hypothesize that different binding patterns might be its chemical structure [208e210]. Monoclonal anti-
induced in vivo as a response to environmental and bodies against B. fragilis LPSs have been shown to
host cues. rescue from peritoneal infections and bacteremia in
nonimmune mice [211].
Response to Oxidative Stress
Gut bacteria are exposed to oxygen outside of the Mechanisms of Interbacterial
host and within the intestinal cavity owing to the
oxygen concentration gradient across the lumen
Competition
[182]. Bacteroides clinical isolates are more aero-
tolerant than nonclinical isolates, suggesting a role T6SS in Bacteroides
for such adaptation in pathogenesis [183]. Oxidative
stress response (OSR) mechanisms might be Bacteria that inhabit the gut are present in large
especially protective in the context of extraintestinal numbers, and competition for a stable place in such
infections, where tissues are more oxygenated than an environment is commonly established [212,213].
the gut lumen [184,185]. The anaerobic/aerobic flux Secretion of toxic proteins is a common mechanism
can modify gene expression profiles, potentially of competition [214e216], perhaps best exemplified
altering pathogenicity of the organism and its through multiple studies that have recently indicated
association with abdominal and peritoneal infections the importance of the T6SS in Bacteroides niche
[186,187]. Although B. fragilis can grow in nanomolar establishment [46e48] and more broadly in inter-
oxygen concentrations [188], exposure to higher bacterial competition within the intestine [217e220].
levels can arrest its growth [189]; hence, OSR is The T6SS resembles an inverted phage, with
required to avoid oxidative cellular damage [184]. sequence and structural analysis indicative of
OSR is achieved by a vast repertoire of proteins with orthology between these systems [221]. The T6SS
different functions, including superoxide dismutase functions by secreting effectors (toxins) directly from
[190], catalase [191], peroxidases [185,192], iron the cytosol of the bacterial cell into another cell in a
storage proteins [193e196], and thioredoxins [197]. contact-dependent manner [222]. Secretion of T6SS
The transcription factor OxyR is responsible for the effectors by a bacterial cell is accompanied by
772 Good gone bad

synthesis of specific immunity proteins that confer property of colonization resistance is strongly
resistance to attack by sister cells. Each effector dependent on the strain and temporal order of
protein contains a cognate immunity protein, typi- colonization of both the NTBF and ETBF strains to
cally encoded by neighboring genes. Although it has be tested [50].
been shown that the T6SS can be used by certain
bacteria to inject toxins into eukaryotic cells, most
Antimicrobial Proteins in Bacteroides
species use the T6SS as a mechanism of inter-
bacterial killing [223]. In addition to contact-dependent killing, Bacter-
In the species of the order Bacteroidales, the oides can secrete soluble toxins. In particular,
genes encoding the T6SS can be present in three Bacteroides genomes carry genes that encode
different genetic arrangements known as genomic toxins that belong to a group named Bacteroi-
architectures (GAs), GA1, GA2, and GA3. While dales-secreted antimicrobial proteins (BSAPs)
GA1 and GA2 are found in many species of the order [51e53]. BSAPs are orthologs of membrane attack
Bacteroidales, GA3 is restricted to B. fragilis [49]. An complex/perforin (MACPF) domainecontaining pro-
evolutionary explanation to B. fragilis GA3 restriction teins and have been shown to be important for
is that GA1 and GA2 can be mobilized between strain competition between Bacteroides species in
bacterial cells through genetic elements, but GA3 vitro and in vivo [51e53]. Bacteroides species can
does not contain such features of transmission [49]. also secrete a ubiquitin-like protein that mediates
T6SS genes from Bacteroides sp. are present in up interbacterial killing [54]. The wide variety of
to 75% of the population as per sequence analysis contact-dependent and contact-independent
from the Human Microbiome Project. This high mechanisms and effector molecules for bacterial
prevalence of T6SS-bearing Bacteroides in humans competition reinforces the concept that a single
suggests that this locus plays an important role in the genetic determinant will not suffice to outcompete
competition for colonic niche establishment. Most many diverse organisms from a given niche. Multi-
adult human hosts are colonized by a single strain of ple genetic factors likely govern niche occupancy
B. fragilis; however, during infancy, strain domi- by B. fragilis; hence, genomic diversity between
nance can shift, evidenced by differential relative strains reduces the possibility of a single strain to be
abundance of E-I pairs over time [14]. This finding used as a general probiotic to displace ETBF from
generates a compelling argument that the T6SS is a the gut ecosystem. It seems clear at this point that
critical feature for initial niche acquisition during early curation of gene function is still one of the major
childhood. The same study shows an association bottlenecks in understanding the role of the
between T6SS-bearing B. fragilis and an increase in 15e20% strain-specific portion of B. fragilis gen-
other species of Bacteroides in the same ecosys- omes in colonization.
tem, suggesting that GA3 effector-immunity pairs are
used in vivo for competition mostly between
B. fragilis strains [14]. This could be due to other Host Factors in Colonization and
Bacteroides species occupying different geographic Disease
niches within the colon, limiting the negative effect of
the T6SS from B. fragilis [47]. In addition, other When B. fragilis initially encounters the colon
Bacteroides species could be less sensitive in vivo to during early infancy, it is immersed in an environ-
B. fragilis T6SS-dependent killing than B. fragilis ment where constant and deep changes are
strains carrying different GA3 E-I pairs [47]. A very occurring to host and microbial cells [225,226].
recent report has shed light on the mechanistic basis Maturation of the colonic epithelial lining and mucus
of coexistence between B. fragilis and other species layer are the key to avoid excessive exposure of the
from the same genus. Many Bacteroides species host to microbes and prevent inflammation
have acquired additional immunity proteins against [227,228]. The main structural component of the
GA3 from B. fragilis, gaining the capacity to defend mucus is the glycoprotein MUC2, secreted by
from GA3 attacks and potentially exploit goblet cells [229]. MUC2 and other mucins form a
B. fragiliseoccupied niches [224]. The role of the dense mucus layer in direct contact with epithelial
T6SS in bacterial competition makes it a potentially cells and a loose mucus layer around 50 mm apart
useful tool to manipulate carriage of pathogens by from the epithelium [230]. The loose mucus layer
probiotic interventions. We have shown that coin- allows penetration by many microorganisms,
oculation and competition between wild-type ETBF whereas the dense layer restricts access to the
(strain 43858) and NTBF (strain NCTC 9343) in epithelium to a small group of bacteria [230,231].
antibiotic-treated mice reduces the burden and Thickness and penetrability of the mucus is highly
toxicity of ETBF in a T6SS-dependent manner. influenced by microbiota composition, highlighting
When NTBF lacks an active T6SS or the effector the complex relationship between the variables
protein from the operon, ETBF is able to colonize affecting gut health [232,233]. Challenge with ETBF
successfully and drive colonic disease [50]. This in Muc2 mucus-deficient mice leads to high
Good gone bad 773

lethality, indicating the key role of the mucus layer in


preventing ETBF-associated disease [96]. Con-
stant and dynamic communication between host
cells and microbiota during intestinal maturation
contributes to achievement of a symbiotic steady
state that can be disrupted by perturbation of either
the host or the microbiome constituents. In this
section, we will focus on the known host determi-
nants for progression of disease caused by ETBF.

Association of ETBF With Chronic Disease in


Humans

The association between colitis and CRC with


carriage of ETBF has led to the hypothesis that this
strain, because of its tissue-damaging toxin, is a
causal agent of CRC instead of a bystander such as
Streptococcus gallolyticus [234]. Most cases of
CRC (~90%) are sporadic whereas a minor
percentage are familial in nature. In both sporadic
and familial CRC, mutations in the adenomatous
polyposis coli (APC) gene are present in 70e80% of
cases [235,236]. The lack of functional APC is one Fig. 4. Effects of BFT on the immune response.
of the main drivers of polyp disease and early-onset Activation of b-catenin/Wnt, STAT3, and NF-kb pathways
CRC [237]. APC forms a protein complex that is required for cell damage and inflammation. Macro-
targets free cytosolic b-catenin to degradation phages within the lamina propria secrete proinflammatory
through ubiquitinylation [237]. When E-cadherin is cytokines that promote a Th17 cellular phenotype. The NF-
cleaved in a BFT-dependent manner, more soluble kB pathway requires IL-17 signaling in CECs. CECs
b-catenin is available for Wnt/b-catenin signaling, secrete CXC chemokines such as IL-8, promoting recruit-
ment of myeloid cells to the site of damage. BFT,
inducing a proliferative response through c-myc
Bacteroides fragilis toxin; CEC, colonic epithelial cell; IL,
(Fig. 3) [105]. Thus, mutations that abrogate interleukin; MAPK, mitogen-activated protein kinase.
functional APC, together with the effect of BFT on
E-cadherin and the free cytosolic pool of b-catenin,
create the conditions for cellular hyperproliferation tease-producing (colibactin) pks and E. coli, restrict-
and crypt hyperplasia. The use of multiple intestinal ing the presence of other microorganisms (Fig. 5)
neoplasia (Min) mouse models carrying mutations [17]. Coinoculation of both bacteria in an azoxy-
in APC has promoted a clearer causal relationship methane-treated mice model of colon cancer
between ETBF and CRC [18,238,239]. These mice increased severity of the disease, dependent on
spontaneously develop tumors in the small intestine ETBF-driven secretion of IL-17 [17]. Most recently,
after 2e3 months in the presence of their endogen- mucosal bacterial communities from both healthy
ous microbiota. However, after oral gavage with individuals and patients with colon cancer were
ETBF, colonic disease appears ~4 weeks after shown to induce tumors in three Apc mouse
inoculation [239]. BFT-driven damage of colonic models, implying that such communities would be
cells in APC-mutant mice triggers a hyperprolifera- contributors to disease in predisposed individuals
tive response (Fig. 3) and inflammatory cascade [240]. Major perturbations of gut epithelial home-
(Fig. 4) in a Stat3- and IL-17edependent manner ostasis seem to be accompanied of more severe
[16,18,239]. Secretion of IL-8 and other CXC ETBF-dependent phenotypes.
chemokines recruits immature polymorphonuclear The intrinsic predisposition of each individual to
cells, leading to exacerbation of inflammation and colonic inflammation and dysbiosis is likely to
cell damage (Fig. 4) [16,18]. These experiments govern whether ETBF displays a phenotype con-
indicate that ETBF can remodel the colonic epithe- sistent with asymptomatic colonization or severe
lium drastically in a predisposed model organism colonic disease. As shown in Fig. 5, host suscept-
toward a state of disease. A very recent study ibility to ETBF is determined by the state of the
looked at colon biopsies from human patients with mammalian host cells per se and the symbiotic
familial adenomatous polyposis and found evi- microorganisms that together form a complex and
dence of causal association between ETBF and dynamic ecosystem. During periods of gut home-
development of CRC [17]. In these individuals, ostasis, either toxin production could be abolished
ETBF was present in biofilms together with pro- or become nondamaging in a host that presents a
774 Good gone bad

Fig. 5. Host and microbial factors that modulate degree of commensalism or pathogenicity of ETBF. ETBF
carriage can result in a risk of colonic disease owing to secretion of BFT and cell damage, but host predisposition is the key
to disease by ETBF. A healthy steady-state microbial community that is not permissive to colonization by ETBF should
contain NTBF bacteria that compete for the colonic niche with ETBF. NTBF requires the capsule to colonize and establishes
competition through the T6SS and BSAPs. In addition, colonization by bacterial commensals during early infancy drives
secretion of anti-inflammatory IL-10, contributing to immune tolerance and microbial symbiosis. On the other hand, damage
caused by BFT and pathogenicity can be worsened by stable colonization by ETBF through bactericidal mechanisms such
as T6SS and BSAPs and specific host susceptibility factors including APC mutations, defects in the epithelial mucus layer,
and colonization by other deleterious microbes such as protease-positive (pksþ) E. coli. BFT, Bacteroides fragilis toxin;
ETBF, enterotoxigenic Bacteroides fragilis; T6SS, type VI secretion system; BSAP, Bacteroidales-secreted antimicrobial
protein; APC, adenomatous polyposis coli; NTBF, non-toxigenic B. fragilis; IL, interleukin.

healthy mucus layer and a symbiotic microbiome. most niche acquisition determinants remain fairly
Perturbations of the gut environment could trigger unexplored [141].
pathogenicity of ETBF, resulting in inflammation
and dysbiosis. Changes in bacterial composition
and host cell metabolism could be fueling the ETBF as Part of a Complex Ecological
ecosystem with metabolites that can be hijacked Scenario
by ETBF, in a similar way to what has been reported
for S.enterica and V.cholerae [55,241]. A case for B. fragilis is one of the least abundant species of
that is polyamine metabolism, affected in mice Bacteroides spp. in humans, comprising around
colonized with ETBF in a BFT-dependent manner. 0.5% of total gut bacteria, yet is the most commonly
Spermine oxidase catalyzes formation of Reactive isolated Bacteroides species from samples of infec-
Oxygen Species (ROS), and its expression is tions. As previously mentioned, human metagen-
increased in HT29 cells exposed to purified BFT, ome data in children show that this period of life is
and in C57BL/6 mice, causing DNA damage and pivotal for initial niche acquisition by B. fragilis and is
inflammation (Fig. 3) [242]. Probiotic interventions characterized by fierce strain competition through
could be a very valuable tool to delay progression of the T6SS [14]. From an evolutionary perspective, it is
disease caused by ETBF in predisposed indivi- tempting to speculate that environmental pressure
duals. It has recently been shown that in Min mice, for niche establishment could have positively
coinoculation of mice with NTBF (strain NCTC selected the presence of additional factors such as
9343) and ETBF (strain 86-5443-2-2, or 86), or BFT that allows ETBF to trespass the colonic
inoculation of NTBF after ETBF colonization, does epithelial barrier and gain access to other niches. A
not rescue the progression of chronic colitis and link between the site of infection by B. fragilis and its
CRC [141]. Therefore, previous niche occupancy by original niche localization in the gut remains to be
ETBF highly reduces the success of a second found.
acquisition event by a probiotic NTBF strain The generation of efficacious probiotic interven-
[50,141]. In this study, dominance of ETBF over tions requires a detailed molecular understanding of
NTBF is partly explained by the secretion of a the repertoire of genetic determinants of niche
MACPF non-T6SS toxin, highlighting how poten- acquisition within many strains of B. fragilis. Geno-
tially complex probiotic interventions would be in mic acquisition of bft has occurred through multiple
clinical settings, where genetic diversity is high, and independent events of horizontal transmission [92].
Good gone bad 775

Thus, diverse genetic backgrounds with different resolution than enumeration of colony-forming
capacities for niche establishment and strain com- units from stool samples to understand the rele-
petition can produce BFT and potentially cause vance of bacterial niche occupancy to human health
disease. A desirable probiotic strain against ETBF and disease. Recent research has clearly shown
should attempt to outcompete various strains by that fecal matter does not necessarily reflect the
using “designed” combinations of genetic determi- abundance and degree of colonization of specific
nants of colonization; given the complexity of host gut mucosal tissues by bacteria [245,246], high-
colonization, these may require a personalized lighting the need for detailed investigation of the
medicine perspective to effectively treat those at microbial niche with a focus on anatomic resolution.
risk of ETBF-associated disease. To this end, The use of new tools that enable analysis of in vivo
knowledge within the field should progress toward regulation of gene expression and visualization of
understanding genetic determinants of colonization Bacteroides cells directly from intestinal tissue
and how these can be strategically manipulated to sections will be crucial to dissect the process of
subvert a given strain of ETBF. Moving toward such host colonization in a temporal, spatial, and genetic
an approach will require genomic analysis of multiple manner [247,248]. A novel observation in humans
human isolates and a comprehensive understanding shows that B. fragilis continues adapting within
of the determinants of niche competition. healthy individuals via de novo mutations. Long-
One of the major challenges to the field is to itudinal sampling from a human cohort has shown
understand the initial niche acquisition interactions the appearance of novel strain variants that coexist
of B. fragilis with the human host and to define how in the same host, increasing our grasp on micro-
these interactions modulate the immune system biome biodiversity within a single individual [249]. In
toward a state of tolerance that promotes niche this review, we have focused on genetic determi-
stability and symbiosis. While these events most nants from the host-pathobiont side of the B. fragilis/
likely occur during infancy, further studies will be ETBF story. There are other important factors for
necessary to understand the kinetics of niche colonization, such as host diet, antibiotics, and
acquisition in the human population. In mouse bacteriophages, that are crucial in the development
models, the time frame for early immunological and stability of the lower Gastrointestinal (GI)
recognition and generation of tolerance to a Bacter- ecosystem. A challenge for the field will therefore
oides antigen spans between 10 and 20 days after be the use of emerging complex data analysis
birth [243]. Nonexposure to gut bacterial antigens platforms to integrate host and microbial factors in a
during that time frame or prolonged exposure temporal manner and to better comprehend how
beyond weaning is detrimental to tolerance by the pathobionts such as ETBF can acquire a niche in
immune system, even to gut bacteria that become the gut, can become a stable member of the
dominant later in life [243]. Longitudinal studies microbiome, and can cause disease progression
involving large cohorts of infants will be necessary to in predisposed hosts.
define ETBF prevalence and to be able to narrow
down the temporal aspect of initial host-pathobiont
interactions [225]. The current lack of such studies is
one of the major caveat in the comprehension of how
early childhood acquisition of ETBF impacts disease
progression in adults. Studies to date in mouse
Acknowledgments
models would suggest that ETBF-driven disease is a This work was supported by the Department of
chronic condition in predisposed individuals, result- Pediatrics at Washington University and by the grant
ing from loss of homeostasis of the colonic cells and R01 AI138565-A1 (to J.B.W.).
microbiota over long periods of time [244]. Hence,
understanding early events in stable niche occu- Received 18 April 2019;
pancy by ETBF is probably pivotal for understanding Received in revised form 27 November 2019;
the risk of progression of disease. We are still Accepted 5 December 2019
unaware of how BFT expression and secretion is Available online 17 December 2019
regulated in vivo in humans. It is unknown whether
low BFT production or toxin resistance by the host is Keywords:
responsible for such a high proportion of asympto- Bacteroides fragilis;
matic carriers of ETBF. The process of long-term BFT;
and stable colonization of humans by B. fragilis Fragilysin;
remains as uncharted territory. Metalloprotease;
The field is now poised with an opportunity to Gut microbiota
develop and use methods with higher spatial
776 Good gone bad

[15] C.L. Sears, S. Islam, A. Saha, M. Arjumand, N.H. Alam,


References A.S.G. Faruque, M.A. Salam, J. Shin, D. Hecht,
A. Weintraub, R.B. Sack, F. Qadri, Association of enter-
otoxigenic Bacteroides fragilis infection with inflammatory
[1] T. Yatsunenko, F.E. Rey, M.J. Manary, I. Trehan, diarrhea, Clin. Infect. Dis. 47 (2008) 797e803, https://
M.G. Dominguez-Bello, M. Contreras, M. Magris, doi.org/10.1086/591130.
G. Hidalgo, R.N. Baldassano, A.P. Anokhin, A.C. Heath, [16] E. Thiele Orberg, H. Fan, A.J. Tam, C.M. Dejea,
B. Warner, J. Reeder, J. Kuczynski, J.G. Caporaso, C.E. Destefano Shields, S. Wu, L. Chung, B.B. Finard,
C.A. Lozupone, C. Lauber, J.C. Clemente, D. Knights, X. Wu, P. Fathi, S. Ganguly, J. Fu, D.M. Pardoll,
R. Knight, J.I. Gordon, Human gut microbiome viewed C.L. Sears, F. Housseau, The myeloid immune signature
across age and geography, Nature 486 (2012) 222, https:// of enterotoxigenic Bacteroides fragilis-induced murine
doi.org/10.1038/nature11053. colon tumorigenesis, Mucosal Immunol. 10 (2017)
[2] C.A. Lozupone, J.I. Stombaugh, J.I. Gordon, J.K. Jansson, 421e433, https://doi.org/10.1038/mi.2016.53.
R. Knight, Diversity, stability and resilience of the human gut [17] C.M. Dejea, P. Fathi, J.M. Craig, A. Boleij, R. Taddese,
microbiota, Nature 489 (2012) 220e230, https://doi.org/ A.L. Geis, X. Wu, C.E. DeStefano Shields,
10.1038/nature11550. E.M. Hechenbleikner, D.L. Huso, R.A. Anders,
[3] J.A. Gilbert, M.J. Blaser, J.G. Caporaso, J.K. Jansson, S. V F.M. Giardiello, E.C. Wick, H. Wang, S. Wu, D.M. Pardoll,
Lynch, R. Knight, Current understanding of the human F. Housseau, C.L. Sears, Patients with familial adenoma-
microbiome, Nat. Med. 24 (2018) 392e400, https://doi.org/ tous polyposis harbor colonic biofilms containing tumori-
10.1038/nm.4517. genic bacteria, Science 359 (2018) 592e597, https://
[4] W.E. Moore, L. V Holdeman, Human fecal flora: the normal doi.org/10.1126/science.aah3648.
flora of 20 Japanese-Hawaiians, Appl. Microbiol. 27 (1974) [18] L. Chung, E. Thiele Orberg, A.L. Geis, J.L. Chan, K. Fu,
961e979. C.E. DeStefano Shields, C.M. Dejea, P. Fathi, J. Chen,
[5] D.A. Leigh, Clinical importance of infections due to B.B. Finard, A.J. Tam, F. McAllister, H. Fan, X. Wu,
Bacteroides fragilis and role of antibiotic therapy, Br. Med. S. Ganguly, A. Lebid, P. Metz, S.W. Van Meerbeke,
J. 3 (1974) 225e228. D.L. Huso, E.C. Wick, D.M. Pardoll, F. Wan, S. Wu,
[6] R.W. Cooke, Bacteroides fragilis septicaemia and meningi- C.L. Sears, F. Housseau, Bacteroides fragilis toxin co-
tis in early infancy, Arch. Dis. Child. 50 (1975) 241e243. ordinates a pro-carcinogenic inflammatory cascade via
[7] D.A. Leigh, Wound infections due to Bacteroides fragilis targeting of colonic epithelial cells, Cell Host Microbe 23
following intestinal surgery, Br. J. Surg. 62 (1975) 375e378. (2018) 203e214, https://doi.org/10.1016/j.chom.
[8] H. Olsen, Bacteroides bacteraemia. A clinical and bacter- 2018.01.007, e5.
iological analysis of 51 patients, Scand. J. Infect. Dis. 8 [19] L.L. Myers, B.D. Firehammer, D.S. Shoop, M.M. Border,
(1976) 107e111. Bacteroides fragilis: a possible cause of acute diarrheal
[9] B. Olsson, C.E. Nord, T. Wadstrom, Formation of beta- disease in newborn lambs, Infect. Immun. 44 (1984) 241e244.
lactamase in Bacteroides fragilis: cell-bound and extracel- [20] L.L. Myers, D.S. Shoop, B.D. Firehammer, M.M. Border,
lular activity, Antimicrob. Agents Chemother. 9 (1976) Association of enterotoxigenic Bacteroides fragilis with
727e735. diarrheal disease in calves, J. Infect. Dis. 152 (1985)
[10] D.J. Blazevic, Antibiotic susceptibility of the subspecies of 1344e1347.
Bacteroides fragilis, Antimicrob. Agents Chemother. 9 [21] L.L. Myers, D.S. Shoop, Association of enterotoxigenic
(1976) 481e484. Bacteroides fragilis with diarrheal disease in young pigs,
[11] J.S. Salaki, R. Black, F.P. Tally, J.W. Kislak, Bacteroides Am. J. Vet. Res. 48 (1987) 774e775, https://doi.org/
fragilis resistant to the administration of clindamycin, Am. J. 10.1016/S0167-2991(08)64389-X.
Med. 60 (1976) 426e428. [22] L.L. Myers, D.S. Shoop, L.L. Stackhouse, F.S. Newman,
[12] T. Vatanen, D.R. Plichta, J. Somani, P.C. Munch, R.J. Flaherty, G.W. Letson, R.B. Sack, Isolation of
T.D. Arthur, A.B. Hall, S. Rudolf, E.J. Oakeley, X. Ke, enterotoxigenic Bacteroides fragilis from humans with
R.A. Young, H.J. Haiser, R. Kolde, M. Yassour, diarrhea, J. Clin. Microbiol. 25 (1987) 2330e2333, https://
K. Luopajarvi, H. Siljander, S.M. Virtanen, J. Ilonen, doi.org/10.1177/0146167293195003.
R. Uibo, V. Tillmann, S. Mokurov, N. Dorshakova, [23] L.L. Myers, D.S. Shoop, T.D. Byars, Diarrhea associated
J.A. Porter, A.C. McHardy, H. Lahdesmaki, H. Vlamakis, with enterotoxigenic Bacteroides fragilis in foals, Am. J. Vet.
C. Huttenhower, M. Knip, R.J. Xavier, Genomic variation Res. 48 (1987) 1565e1567.
and strain-specific functional adaptation in the human gut [24] D.S. Shoop, L.L. Myers, J.B. LeFever, Enumeration of
microbiome during early life, Nat. Microbiol. 4 (2019) enterotoxigenic Bacteroides fragilis in municipal sewage,
470e479, https://doi.org/10.1038/s41564-018-0321-5. Appl. Environ. Microbiol. 56 (1990) 2243e2244.
[13] J. Lloyd-Price, A. Mahurkar, G. Rahnavard, J. Crabtree, [25] R.B. Sack, L.L. Myers, J. Almeido-Hill, D.S. Shoop,
J. Orvis, A.B. Hall, A. Brady, H.H. Creasy, C. McCracken, W.C. Bradbury, R. Reid, M. Santosham, Enterotoxigenic
M.G. Giglio, D. McDonald, E.A. Franzosa, R. Knight, Bacteroides fragilis: epidemiologic studies of its role as a
O. White, C. Huttenhower, Strains, functions and dynamics human diarrhoeal pathogen, J. Diarrhoeal Dis. Res. 10
in the expanded human microbiome project, Nature 550 (1992) 4e9, https://doi.org/10.1126/science.1184944.
(2017) 61, https://doi.org/10.1038/nature23889. [26] R.B. Sack, M.J. Albert, K. Alam, P.K. Neogi, M.S. Akbar,
[14] A.J. Verster, B.D. Ross, M.C. Radey, Y. Bao, Isolation of enterotoxigenic Bacteroides fragilis from Ban-
A.L. Goodman, J.D. Mougous, E. Borenstein, The land- gladeshi children with diarrhea: a controlled study, J. Clin.
scape of type VI secretion across human gut microbiomes Microbiol. 32 (1994) 960e963.
reveals its role in community composition, Cell Host [27] G. Zhang, B. Svenungsson, A. Karnell, A. Weintraub,
Microbe 22 (2017) 411e419, https://doi.org/10.1016/ Prevalence of enterotoxigenic Bacteroides fragilis in adult
j.chom.2017.08.010, e4.
Good gone bad 777

patients with diarrhea and healthy controls, Clin. Infect. Dis. ectal neoplasia, PLoS One 12 (2017), e0171602, https://
29 (1999) 590e594. doi.org/10.1371/journal.pone.0171602.
[28] V.H. San Joaquin, J.C. Griffis, C. Lee, C.L. Sears, [41] F. Haghi, E. Goli, B. Mirzaei, H. Zeighami, The association
Association of bacteroides fragilis with childhood diarrhea, between fecal enterotoxigenic B. fragilis with colorectal
Scand. J. Infect. Dis. 27 (1995) 211e215, https://doi.org/ cancer, BMC Canc. 19 (2019) 879, https://doi.org/10.1186/
10.3109/00365549509019011. s12885-019-6115-1.
[29] S.K. Niyogi, P. Dutta, U. Mitra, D.K. Pal, Association of [42] A. Pantosti, M.G. Menozzi, A. Frate, L. Sanfilippo,
enterotoxigenic Bacteroides fragilis with childhood diar- F. D'Ambrosio, M. Malpeli, Detection of enterotoxigenic
rhoea, Indian J. Med. Res. 105 (1997) 167e169. Bacteroides fragilis and its toxin in stool samples from
[30] P. Pathela, K.Z. Hasan, E. Roy, K. Alam, F. Huq, adults and children in Italy, Clin. Infect. Dis. 24 (1997)
A.K. Siddique, R.B. Sack, Enterotoxigenic Bacteroides 12e16, https://doi.org/10.1093/clinids/24.1.12.
fragilis-associated diarrhea in children 0-2 years of age in [43] J.L. Round, S.K. Mazmanian, Inducible Foxp3þ regulatory
rural Bangladesh, J. Infect. Dis. 191 (2005) 1245e1252, T-cell development by a commensal bacterium of the
https://doi.org/10.1086/428947. intestinal microbiota, Proc. Natl. Acad. Sci. U. S. A 107
[31] T. Vu Nguyen, P. Le Van, C. Le Huy, A. Weintraub, ( 2 0 1 0 ) 1 2 2 0 4 e1 2 2 0 9 , h t t p s : / / d o i . o r g / 1 0 . 1 0 7 3 /
Diarrhea caused by enterotoxigenic Bacteroides fragilis in pnas.0909122107.
children less than 5 years of age in Hanoi, Vietnam., [44] J.L. Round, S.M. Lee, J. Li, G. Tran, B. Jabri, T.A. Chatila,
Anaerobe. 11 (2005) 109e114, https://doi.org/10.1016/ S.K. Mazmanian, The Toll-like receptor 2 pathway estab-
j.anaerobe.2004.10.004. lishes colonization by a commensal of the human micro-
[32] B. Durmaz, M. Dalgalar, R. Durmaz, Prevalence of biota, Science 332 (2011) 974e977, https://doi.org/
enterotoxigenic Bacteroides fragilis in patients with diar- 10.1126/science.1206095.
rhea: a controlled study, Anaerobe 11 (2005) 318e321, [45] B.W. Casterline, A.L. Hecht, V.M. Choi, J. Bubeck Warden-
https://doi.org/10.1016/j.anaerobe.2005.06.001. burg, The Bacteroides fragilis pathogenicity island links
[33] T.P. Prindiville, R.A. Sheikh, S.H. Cohen, Y.J. Tang, virulence and strain competition, Gut Microb. 8 (2017)
M.C. Cantrell, J.J. Silva, Bacteroides fragilis enterotoxin 374e383, https://doi.org/10.1080/19490976.2017.1290758.
gene sequences in patients with inflammatory bowel [46] A.B. Russell, A.G. Wexler, B.N. Harding, J.C. Whitney,
disease, Emerg. Infect. Dis. 6 (2000) 171e174, https:// A.J. Bohn, Y.A. Goo, B.Q. Tran, N.A. Barry, H. Zheng,
doi.org/10.3201/eid0602.000210. S.B. Peterson, S. Chou, T. Gonen, D.R. Goodlett,
[34] N.U. Toprak, A. Yagci, B.M. Gulluoglu, M.L. Akin, A.L. Goodman, J.D. Mougous, A type VI secretion-related
P. Demirkalem, T. Celenk, G. Soyletir, A possible role of pathway in Bacteroidetes mediates interbacterial antagon-
Bacteroides fragilis enterotoxin in the aetiology of color- ism, Cell Host Microbe 16 (2014) 227e236, https://doi.org/
ectal cancer, Clin. Microbiol. Infect. 12 (2006) 782e786, 10.1016/j.chom.2014.07.007.
https://doi.org/10.1111/j.1469-0691.2006.01494.x. [47] A.G. Wexler, Y. Bao, J.C. Whitney, L.-M. Bobay,
[35] A. Boleij, E.M. Hechenbleikner, A.C. Goodwin, R. Badani, J.B. Xavier, W.B. Schofield, N.A. Barry, A.B. Russell,
E.M. Stein, M.G. Lazarev, B. Ellis, K.C. Carroll, E. Albesiano, B.Q. Tran, Y.A. Goo, D.R. Goodlett, H. Ochman,
E.C. Wick, E.A. Platz, D.M. Pardoll, C.L. Sears, The J.D. Mougous, A.L. Goodman, Human symbionts inject
Bacteroides fragilis toxin gene is prevalent in the colon and neutralize antibacterial toxins to persist in the gut,
mucosa of colorectal cancer patients, Clin. Infect. Dis. 60 Proc. Natl. Acad. Sci. U. S. A 113 (2016) 3639e3644,
(2015) 208e215, https://doi.org/10.1093/cid/ciu787. https://doi.org/10.1073/pnas.1525637113.
[36] K.S. Viljoen, A. Dakshinamurthy, P. Goldberg, [48] M. Chatzidaki-Livanis, N. Geva-Zatorsky, L.E. Comstock,
J.M. Blackburn, Quantitative profiling of colorectal can- Bacteroides fragilis type VI secretion systems use novel
cer-associated bacteria reveals associations between effector and immunity proteins to antagonize human gut
fusobacterium spp., enterotoxigenic Bacteroides fragilis Bacteroidales species, Proc. Natl. Acad. Sci. U. S. A 113
(ETBF) and clinicopathological features of colorectal ( 2 0 1 6 ) 3 6 2 7 e3 6 3 2 , h t t p s : / / d o i . o r g / 1 0 . 1 0 7 3 /
cancer, PLoS One 10 (2015), e0119462, https://doi.org/ pnas.1522510113.
10.1371/journal.pone.0119462. [49] M.J. Coyne, K.G. Roelofs, L.E. Comstock, Type VI
[37] Y. Zhou, H. He, H. Xu, Y. Li, Z. Li, Y. Du, J. He, Y. Zhou, secretion systems of human gut Bacteroidales segregate
H. Wang, Y. Nie, Association of oncogenic bacteria with into three genetic architectures, two of which are contained
colorectal cancer in South China, Oncotarget 7 (2016) on mobile genetic elements, BMC Genomics 17 (2016) 58,
80794e80802, https://doi.org/10.18632/oncotarget.13094. https://doi.org/10.1186/s12864-016-2377-z.
[38] J.I. Keenan, A. Aitchison, R. V Purcell, R. Greenlees, [50] A.L. Hecht, B.W. Casterline, Z.M. Earley, Y.A. Goo,
J.F. Pearson, F.A. Frizelle, Screening for enterotoxigenic D.R. Goodlett, J. Bubeck Wardenburg, Strain competition
Bacteroides fragilis in stool samples, Anaerobe 40 (2016) restricts colonization of an enteric pathogen and prevents
50e53, https://doi.org/10.1016/j.anaerobe.2016.05.004. colitis, EMBO Rep. 17 (2016) 1281e1291, https://doi.org/
[39] S. Zamani, S. Hesam Shariati, M.R. Zali, H. Asadzadeh 10.15252/embr.201642282.
Aghdaei, A. Sarabi Asiabar, S. Bokaie, B. Nomanpour, [51] M. Chatzidaki-Livanis, M.J. Coyne, L.E. Comstock, An
L.A. Sechi, M.M. Feizabadi, Detection of enterotoxigenic antimicrobial protein of the gut symbiont Bacteroides fragilis
Bacteroides fragilis in patients with ulcerative colitis, Gut with a MACPF domain of host immune proteins, Mol.
Pathog. 9 (2017) 53, https://doi.org/10.1186/s13099-017- Microbiol. 94 (2014) 1361e1374, https://doi.org/10.1111/
0202-0. mmi.12839.
[40] R. V Purcell, J. Pearson, A. Aitchison, L. Dixon, [52] K.G. Roelofs, M.J. Coyne, R.R. Gentyala, M. Chatzidaki-
F.A. Frizelle, J.I. Keenan, Colonization with enterotoxigenic Livanis, L.E. Comstock, Bacteroidales secreted antimicro-
Bacteroides fragilis is associated with early-stage color- bial proteins target surface molecules necessary for gut
778 Good gone bad

colonization and mediate competition in vivo, mBio 7 [67] V.M. Choi, J. Herrou, A.L. Hecht, W.P. Teoh, J.R. Turner,
(2016), https://doi.org/10.1128/mBio.01055-16. S. Crosson, J. Bubeck Wardenburg, Activation of Bacter-
[53] A.M. Shumaker, V.L. McEneany, M.J. Coyne, P.A. Silver, oides fragilis toxin by a novel bacterial protease contributes
L.E. Comstock, Identification of a fifth antibacterial toxin to anaerobic sepsis in mice, Nat. Med. 22 (2016) 563e567,
produced by a single Bacteroides fragilis strain, J. Bacteriol. https://doi.org/10.1038/nm.4077.
(2019), https://doi.org/10.1128/JB.00577-18. [68] A.B. Onderdonk, D.L. Kasper, R.L. Cisneros, J.G. Bartlett,
[54] M. Chatzidaki-Livanis, M.J. Coyne, K.G. Roelofs, The capsular polysaccharide of Bacteroides fragilis as a
R.R. Gentyala, J.M. Caldwell, L.E. Comstock, Gut sym- virulence factor: comparison of the pathogenic potential of
biont Bacteroides fragilis secretes a eukaryotic-like ubiqui- encapsulated and unencapsulated strains, J. Infect. Dis.
tin protein that mediates intraspecies antagonism, mBio 8 136 (1977) 82e89, https://doi.org/10.1093/infdis/136.1.82.
(2017), https://doi.org/10.1128/mBio.01902-17. [69] C.S. Weikel, F.D. Grieco, J. Reuben, L.L. Myers, R.B. Sack,
[55] F. Rivera-Chavez, J.J. Mekalanos, Cholera toxin promotes Human colonic epithelial cells, HT29/C1, treated with crude
pathogen acquisition of host-derived nutrients, Nature 572 Bacteroides fragilis enterotoxin dramatically alter their
(2019) 244e248, https://doi.org/10.1038/s41586-019-1453- morphology, Infect. Immun. 60 (1991) 321e327.
3. [70] R.L. Van Tassell, D.M. Lyerly, T.D. Wilkins, Purification and
[56] M. Diard, W.-D. Hardt, Basic processes in salmonella-host characterization of an enterotoxin from Bacteroides fragilis,
interactions: within-host evolution and the transmission of Infect. Immun. 60 (1992) 1343e1350.
the virulent genotype, Microbiol. Spectr. 5 (2017), https:// [71] J.S. Moncrief, R. Obiso, L.A. Barroso, J.J. Kling,
doi.org/10.1128/microbiolspec.MTBP-0012-2016. R.L. Wright, R.L. Van Tassell, D.M. Lyerly, T.D. Wilkins,
[57] J.G. Conner, J.K. Teschler, C.J. Jones, F.H. Yildiz, Staying The enterotoxin of Bacteroides fragilis is a metalloprotease,
alive: Vibrio cholerae's cycle of environmental survival, Infect. Immun. 63 (1995) 175e181.
transmission, and dissemination, Microbiol. Spectr. 4 [72] R.L. Van Tassell, D.M. Lyerly, T.D. Wilkins, Characteriza-
(2016), https://doi.org/10.1128/microbiolspec.VMBF-0015- tion of enterotoxigenic Bacteroides fragilis by a toxin-
2015. specific enzyme-linked immunosorbent assay, Clin. Diagn.
[58] J. Papaparaskevas, A. Katsandri, A. Pantazatou, I. Stefanou, Lab. Immunol. 1 (1994) 578e584.
A. Avlamis, N.J. Legakis, A. Tsakris, Epidemiological char- [73] S. Mootien, P.M. Kaplan, Monoclonal antibodies specific for
acteristics of infections caused by Bacteroides, Prevotella and Bacteroides fragilis enterotoxins BFT1 and BFT2 and their
Fusobacterium species: a prospective observational study, use in immunoassays, PLoS One 12 (2017), e0173128,
Anaerobe 17 (2011) 113e117, https://doi.org/10.1016/ https://doi.org/10.1371/journal.pone.0173128.
J.ANAEROBE.2011.05.013. [74] J.J. Kling, R.L. Wright, J.S. Moncrief, T.D. Wilkins, Cloning
[59] A. Pantosti, M. Cerquetti, R. Colangeli, F. D'Ambrosio, and characterization of the gene for the metalloprotease
Detection of intestinal and extra-intestinal strains of enterotoxin of Bacteroides fragilis, FEMS Microbiol. Lett.
enterotoxigenic Bacteroides fragilis by the HT-29 cytotoxi- 146 (1997) 279e284, https://doi.org/10.1111/j.1574-
city assay, J. Med. Microbiol. 41 (1994) 191e196, https:// 6968.1997.tb10205.x.
doi.org/10.1099/00222615-41-3-191. [75] A.A. Franco, L.M. Mundy, M. Trucksis, S. Wu, J.B. Kaper,
[60] N. Kato, A. Karuniawati, R. Jotwani, H. Kato, K. Watanabe, C.L. Sears, Cloning and characterization of the Bacteroides
K. Ueno, Isolation of enterotoxigenic Bacteroides fragilis fragilis metalloprotease toxin gene, Infect. Immun. 65
from extraintestinal sites by cell culture assay, Clin. Infect. (1997) 1007e1013.
Dis. 20 (Suppl 2) (1995) S141. [76] A.A. Franco, S.L. Buckwold, J.W. Shin, M. Ascon, C.L. Sears,
[61] L.M. Mundy, C.L. Sears, Detection of toxin production by Mutation of the zinc-binding metalloprotease motif affects
Bacteroides fragilis: assay development and screening of Bacteroides fragilis toxin activity but does not affect propeptide
extraintestinal clinical isolates, Clin. Infect. Dis. 23 (1996) processing, Infect. Immun. 73 (2005) 5273e5277, https://
269e276. doi.org/10.1128/IAI.73.8.5273-5277.2005.
[62] G.T. Chung, A.A. Franco, S. Wu, G.E. Rhie, R. Cheng, [77] C.L. Sears, S.L. Buckwold, J.W. Shin, A.A. Franco, The C-
H.B. Oh, C.L. Sears, Identification of a third metallopro- terminal region of Bacteroides fragilis toxin is essential to its
tease toxin gene in extraintestinal isolates of Bacteroides biological activity, Infect. Immun. 74 (2006) 5595e5601,
fragilis, Infect. Immun. 67 (1999) 4945e4949. https://doi.org/10.1128/IAI.00135-06.
[63] M.C. Claros, Z.C. Claros, D.W. Hecht, D.M. Citron, [78] T. Goulas, J.L. Arolas, F.X. Gomis-Rüth, Structure, function
E.J.C. Goldstein, J.J. Silva, Y. Tang-Feldman, and latency regulation of a bacterial enterotoxin potentially
A.C. Rodloff, Characterization of the Bacteroides fragilis derived from a mammalian adamalysin/ADAM xenolog,
pathogenicity island in human blood culture isolates, Proc. Natl. Acad. Sci. 108 (2011) 1856, https://doi.org/
Anaerobe 12 (2006) 17e22, https://doi.org/10.1016/j.anae- 10.1073/pnas.1012173108. LP e 1861.
robe.2005.06.005. [79] J. Herrou, V.M. Choi, J. Bubeck Wardenburg, S. Crosson,
[64] I. Foulon, D. Pierard, G. Muyldermans, K. Vandoorslaer, Activation mechanism of the Bacteroides fragilis cysteine
O. Soetens, P. Rosseel, S. Lauwers, Prevalence of peptidase, fragipain, Biochemistry 55 (2016) 4077e4084,
fragilysin gene in Bacteroides fragilis isolates from blood https://doi.org/10.1021/acs.biochem.6b00546.
and other extraintestinal samples, J. Clin. Microbiol. 41 [80] J.S. Moncrief, A.J. Duncan, R.L. Wright, L.A. Barroso,
(2003) 4428e4430. T.D. Wilkins, Molecular characterization of the fragilysin
[65] N. Polanco, L. Manzi, O. Carmona, [Possible role of pathogenicity islet of enterotoxigenic Bacteroides fragilis,
enterotoxigenic Bacteroides fragilis in the etiology of Infect. Immun. 66 (1998) 1735e1739.
infectious vaginitis], Investig. Clin. 53 (2012) 28e37. [81] A.A. Franco, R.K. Cheng, G.T. Chung, S. Wu, H.B. Oh,
[66] R.J.J. Obiso, A.O. Azghani, T.D. Wilkins, The Bacteroides C.L. Sears, Molecular evolution of the pathogenicity island
fragilis toxin fragilysin disrupts the paracellular barrier of of enterotoxigenic Bacteroides fragilis strains, J. Bacteriol.
epithelial cells, Infect. Immun. 65 (1997) 1431e1439. 181 (1999) 6623e6633, 0021-9193/99/$04.00?0.
Good gone bad 779

[82] N. Kato, C.X. Liu, H. Kato, K. Watanabe, Y. Tanaka, lethal disease, Cell Host Microbe 22 (2017) 443e448,
T. Yamamoto, K. Suzuki, K. Ueno, A new subtype of the https://doi.org/10.1016/j.chom.2017.08.007, e5.
metalloprotease toxin gene and the incidence of the three [97] S.S. Koshy, M.H. Montrose, C.L. Sears, Human intestinal
bft subtypes among Bacteroides fragilis isolates in Japan, epithelial cells swell and demonstrate actin rearrangement
FEMS Microbiol. Lett. 182 (2000) 171e176, https://doi.org/ in response to the metalloprotease toxin of Bacteroides
10.1111/j.1574-6968.2000.tb08892.x. fragilis, Infect. Immun. 64 (1996) 5022e5028.
[83] S. Wu, L.A. Dreyfus, A.O. Tzianabos, C. Hayashi, [98] R.F. Saidi, K. Jaeger, M.H. Montrose, S. Wu, C.L. Sears,
C.L. Sears, Diversity of the metalloprotease toxin produced Bacteroides fragilis toxin rearranges the actin cytoskeleton
by enterotoxigenic Bacteroides fragilis, Infect. Immun. 70 of HT29/C1 cells without direct proteolysis of actin or
(2002) 2463e2471. decrease in F-actin content, Cell Motil. Cytoskelet. 37
[84] A.S. Scotto d'Abusco, M. Del Grosso, S. Censini, (1997) 159e165, https://doi.org/10.1002/(SICI)1097-0169,
A. Covacci, A. Pantosti, The alleles of the bft gene are 1997)37:2<159::AID-CM8>3.0.CO;2-3.
distributed differently among enterotoxigenic Bacteroides [99] S. Wu, K.C. Lim, J. Huang, R.F. Saidi, C.L. Sears,
fragilis strains from human sources and can be present in Bacteroides fragilis enterotoxin cleaves the zonula adhe-
double copies, J. Clin. Microbiol. 38 (2000) 607e612. rens protein, E-cadherin. Proc. Natl. Acad. Sci. U. S. A 95
[85] C.L. Sears, The toxins of Bacteroides fragilis, Toxicon 39 (1998) 14979e14984.
(2001) 1737e1746. [100] S. Wu, J. Shin, G. Zhang, M. Cohen, A. Franco, C.L. Sears,
[86] V. Nakano, D.A. Gomes, R.M.E. Arantes, J.R. Nicoli, The Bacteroides fragilis toxin binds to a specific intestinal
M.J. Avila-Campos, Evaluation of the pathogenicity of the epithelial cell receptor, Infect. Immun. 74 (2006)
Bacteroides fragilis toxin gene subtypes in gnotobiotic 5382e5390, https://doi.org/10.1128/IAI.00060-06.
mice, Curr. Microbiol. 53 (2006) 113e117, https://doi.org/ [101] D. Kharlampieva, V. Manuvera, O. Podgorny, E. Grafskaia,
10.1007/s00284-005-0321-6. S. Kovalchuk, O. Pobeguts, I. Altukhov, V. Govorun,
[87] N. Ulger Toprak, D. Rajendram, A. Yagci, S. Gharbia, V. Lazarev, Recombinant fragilysin isoforms cause E-
H.N. Shah, B.M. Gulluoglu, L.M. Akin, P. Demirkalem, cadherin cleavage of intact cells and do not cleave isolated
T. Celenk, G. Soyletir, The distribution of the bft alleles among E-cadherin, Microb. Pathog. 83e84 (2015) 47e56, https://
enterotoxigenic Bacteroides fragilis strains from stool speci- doi.org/10.1016/j.micpath.2015.05.003.
mens and extraintestinal sites, Anaerobe 12 (2006) 71e74, [102] M. Riegler, M. Lotz, C. Sears, C. Pothoulakis, I. Castagliuolo,
https://doi.org/10.1016/j.anaerobe.2005.11.001. C.C. Wang, R. Sedivy, T. Sogukoglu, E. Cosentini, G. Bischof,
[88] K.P. Sarvari, J. Soki, M. Ivan, C. Miszti, K. Latkoczy, W. Feil, B. Teleky, G. Hamilton, J.T. LaMont, E. Wenzl,
S.Z. Melegh, E. Urban, Detection of enterotoxin and Bacteroides fragilis toxin 2 damages human colonic mucosa
protease genes among Hungarian clinical Bacteroides in vitro, Gut 44 (1999) 504, https://doi.org/10.1136/
fragilis isolates, Anaerobe 48 (2017) 98e102, https:// gut.44.4.504. LP e 510.
doi.org/10.1016/j.anaerobe.2017.07.005. [103] S. Wu, K.-J. Rhee, M. Zhang, A. Franco, C.L. Sears,
[89] A.A. Franco, The Bacteroides fragilis pathogenicity island is Bacteroides fragilis toxin stimulates intestinal epithelial cell
contained in a putative novel conjugative transposon, shedding and gamma-secretase-dependent E-cadherin
J. Bacteriol. 186 (2004) 6077e6092, https://doi.org/ cleavage, J. Cell Sci. 120 (2007) 1944e1952, https://
10.1128/JB.186.18.6077-6092.2004. doi.org/10.1242/jcs.03455.
[90] S.L. Buckwold, N.B. Shoemaker, C.L. Sears, A.A. Franco, [104] S.H.M. Wong, C.M. Fang, L.-H. Chuah, C.O. Leong,
Identification and characterization of conjugative transpo- S.C. Ngai, E-cadherin: its dysregulation in carcinogenesis
sons CTn86 and CTn9343 in Bacteroides fragilis strains, and clinical implications, Crit. Rev. Oncol. Hematol. 121
Appl. Environ. Microbiol. 73 (2007) 53e63, https://doi.org/ ( 2 0 1 8 ) 1 1 e2 2 , h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 / j . c r i t r e -
10.1128/AEM.01669-06. vonc.2017.11.010.
[91] J. Soki, Z. Eitel, G. Terhes, E. Nagy, E. Urban, Occurrence [105] S. Wu, P.J. Morin, D. Maouyo, C.L. Sears, Bacteroides
and analysis of rare cfiA-bft doubly positive Bacteroides fragilis enterotoxin induces c-Myc expression and cellular
fragilis strains, Anaerobe 23 (2013) 70e73, https://doi.org/ proliferation, Gastroenterology 124 (2003) 392e400,
10.1016/j.anaerobe.2013.06.008. https://doi.org/10.1053/gast.2003.50047.
[92] J. V Pierce, H.D. Bernstein, Genomic diversity of enterotoxi- [106] L. Sanfilippo, C.K. Li, R. Seth, T.J. Balwin, M.G. Menozzi,
genic strains of Bacteroides fragilis, PLoS One 11 (2016), Y.R. Mahida, Bacteroides fragilis enterotoxin induces the
e0158171, https://doi.org/10.1371/journal.pone.0158171. expression of IL-8 and transforming growth factor-beta
[93] P. Obuch-Woszczatynski, R.G.F. Wintermans, A. Van (TGF-beta) by human colonic epithelial cells, Clin. Exp.
Belkum, H. Endtz, H. Pituch, D. Kreft, F. Meisel-Mikolajc- Immunol. 119 (2000) 456e463.
zyk, M. Luczak, Enterotoxigenic Bacteroides fragilis (ETBF) [107] J.M. Kim, Y.K. Oh, Y.J. Kim, H.B. Oh, Y.J. Cho, Polarized
strains isolated in The Netherlands and Poland are secretion of CXC chemokines by human intestinal epithelial
genetically diverse, Pol. J. Microbiol. 53 (2004) 35e39. cells in response to Bacteroides fragilis enterotoxin: NF-
[94] A.A. Franco, R.K. Cheng, A. Goodman, C.L. Sears, kappa B plays a major role in the regulation of IL-8
Modulation of bft expression by the Bacteroides fragilis expression, Clin. Exp. Immunol. 123 (2001) 421e427.
pathogenicity island and its flanking region, Mol. Microbiol. [108] J.M. Kim, S.J. Cho, Y.-K. Oh, H.-Y. Jung, Y.-J. Kim, N. Kim,
45 (2002) 1067e1077. Nuclear factor-kappa B activation pathway in intestinal
[95] E.A. Groisman, Feedback control of two-component reg- epithelial cells is a major regulator of chemokine gene
ulatory systems, Annu. Rev. Microbiol. 70 (2016) 103e124, expression and neutrophil migration induced by Bacter-
https://doi.org/10.1146/annurev-micro-102215-095331. oides fragilis enterotoxin, Clin. Exp. Immunol. 130 (2002)
[96] A.L. Hecht, B.W. Casterline, V.M. Choi, J. Bubeck Warden- 59e66.
burg, A two-component system regulates Bacteroides [109] S. Wu, J. Powell, N. Mathioudakis, S. Kane, E. Fernandez,
fragilis toxin to maintain intestinal homeostasis and prevent C.L. Sears, Bacteroides fragilis enterotoxin induces intest-
780 Good gone bad

inal epithelial cell secretion of interleukin-8 through mito- wild-type C57BL/6 mice, Infect. Immun. 77 (2009)
gen-activated protein kinases and a tyrosine kinase- 1708e1718, https://doi.org/10.1128/IAI.00814-08.
regulated nuclear factor-kappaB pathway, Infect. Immun. [121] E.C. Wick, S. Rabizadeh, E. Albesiano, X. Wu, S. Wu,
7 2 ( 2 00 4) 5 83 2e5 83 9, https://doi.org/10.1128/ J. Chan, K.-J. Rhee, G. Ortega, D.L. Huso, D. Pardoll,
IAI.72.10.5832-5839.2004. F. Housseau, C.L. Sears, Stat3 activation in murine colitis
[110] J.M. Kim, H.Y. Jung, J.Y. Lee, J. Youn, C.-H. Lee, K.- induced by enterotoxigenic Bacteroides fragilis, Inflamm.
H. Kim, Mitogen-activated protein kinase and activator Bowel Dis. 20 (2014) 821e834, https://doi.org/10.1097/
protein-1 dependent signals are essential for Bacteroides MIB.0000000000000019.
fragilis enterotoxin-induced enteritis, Eur. J. Immunol. 35 [122] T. Bowman, M.A. Broome, D. Sinibaldi, W. Wharton,
(2005) 2648e2657, https://doi.org/10.1002/eji.200526321. W.J. Pledger, J.M. Sedivy, R. Irby, T. Yeatman,
[111] S. Hwang, S.-Y. Gwon, M.S. Kim, S. Lee, K.-J. Rhee, S.A. Courtneidge, R. Jove, Stat3-mediated Myc expression
Bacteroides fragilis toxin induces IL-8 secretion in HT29/C1 is required for Src transformation and PDGF-induced
cells through disruption of E-cadherin junctions, Immune mitogenesis, Proc. Natl. Acad. Sci. 98 (2001) 7319,
Netw 13 (2013) 213e217, https://doi.org/10.4110/ https://doi.org/10.1073/pnas.131568898. LP e 7324.
in.2013.13.5.213. [123] G.S. Yochum, C.M. Sherrick, M. MacPartlin, R.H. Goodman,
[112] J.M. Kim, J.Y. Lee, Y.M. Yoon, Y.-K. Oh, J.S. Kang, Y.- A b-catenin/TCF-coordinated chromatin loop at <em>MYC</
J. Kim, K.-H. Kim, Bacteroides fragilis enterotoxin induces em> integrates 50 and 30 Wnt responsive enhancers, Proc.
cyclooxygenase-2 and fluid secretion in intestinal epithelial Natl. Acad. Sci. 107 (2010) 145, https://doi.org/10.1073/
cells through NF-kappaB activation, Eur. J. Immunol. 36 pnas.0912294107. LP e 150.
(2006) 2446e2456, https://doi.org/10.1002/eji.200535808. [124] M. Yassour, T. Vatanen, H. Siljander, A.-M. Hamalainen,
[113] J.M. Kim, J.Y. Lee, Y.-J. Kim, Inhibition of apoptosis in T. Harkonen, S.J. Ryhanen, E.A. Franzosa, H. Vlamakis,
Bacteroides fragilis enterotoxin-stimulated intestinal epithe- C. Huttenhower, D. Gevers, E.S. Lander, M. Knip,
lial cells through the induction of c-IAP-2, Eur. J. Immunol. R.J. Xavier, Natural history of the infant gut microbiome
3 8 ( 2 00 8) 2 19 0e2 19 9, https://doi.org/10.1002/ and impact of antibiotic treatment on bacterial strain
eji.200838191. diversity and stability, Sci. Transl. Med. 8 (2016) 343ra81,
[114] S.H. Ko, D.J. Rho, J.I. Jeon, Y.-J. Kim, H.A. Woo, Y.K. Lee, https://doi.org/10.1126/scitranslmed.aad0917.
J.M. Kim, Bacteroides fragilis enterotoxin upregulates [125] D. Rothschild, O. Weissbrod, E. Barkan, A. Kurilshikov,
heme oxygenase-1 in intestinal epithelial cells via a T. Korem, D. Zeevi, P.I. Costea, A. Godneva, I.N. Kalka,
mitogen-activated protein kinase- and NF-kappaB-Depen- N. Bar, S. Shilo, D. Lador, A.V. Vila, N. Zmora, M. Pevsner-
dent pathway, leading to modulation of apoptosis, Infect. Fischer, D. Israeli, N. Kosower, G. Malka, B.C. Wolf,
Immun. 84 (2016) 2541e2554, https://doi.org/10.1128/ T. Avnit-Sagi, M. Lotan-Pompan, A. Weinberger,
IAI.00191-16. Z. Halpern, S. Carmi, J. Fu, C. Wijmenga, A. Zhernakova,
[115] Y.M. Yoon, J.Y. Lee, D. Yoo, Y.-S. Sim, Y.-J. Kim, Y.- E. Elinav, E. Segal, Environment dominates over host
K. Oh, J.S. Kang, S. Kim, J.S. Kim, J.M. Kim, Bacteroides genetics in shaping human gut microbiota, Nature 555
fragilis enterotoxin induces human beta-defensin-2 expres- (2018) 210, https://doi.org/10.1038/nature25973.
sion in intestinal epithelial cells via a mitogen-activated [126] D.L. Kasper, The polysaccharide capsule of Bacteroides
protein kinase/I kappaB kinase/NF-kappaB-dependent fragilis subspecies fragilis: immunochemical and morpho-
pathway, Infect. Immun. 78 (2010) 2024e2033, https:// logic definition, J. Infect. Dis. 133 (1976) 79e87.
doi.org/10.1128/IAI.00118-10. [127] D.L. Kasper, M.E. Hayes, B.G. Reinap, F.O. Craft,
[116] D.Y. Yoo, S.H. Ko, J. Jung, Y.-J. Kim, J.S. Kim, J.M. Kim, A.B. Onderdonk, B.F. Polk, Isolation and identification of
Bacteroides fragilis enterotoxin upregulates lipocalin-2 encapsulated strains of Bacteroides fragilis, J. Infect. Dis.
expression in intestinal epithelial cells, Lab. Investig. 93 136 (1977) 75e81.
(2013) 384e396, https://doi.org/10.1038/labinvest.2013.1. [128] A.A. Lindberg, P. Berthold, C.E. Nord, A. Weintraub,
[117] S.H. Ko, J.I. Jeon, H.S. Myung, Y.-J. Kim, J.M. Kim, Encapsulated strains of Bacteroides fragilis in clinical
Bacteroides fragilis enterotoxin induces formation of specimens, Med. Microbiol. Immunol. 167 (1979) 29e36.
autophagosomes in endothelial cells but interferes with [129] D.L. Kasper, A. Weintraub, A.A. Lindberg, J. Lonngren,
fusion with lysosomes for complete autophagic flux through Capsular polysaccharides and lipopolysaccharides from
a mitogen-activated protein kinase-, AP-1-, and C/EBP two Bacteroides fragilis reference strains: chemical and
homologous protein-dependent pat, Infect. Immun. 85 immunochemical characterization, J. Bacteriol. 153 (1983)
(2017), https://doi.org/10.1128/IAI.00420-17. 991e997.
[118] J. Allen, S. Hao, C.L. Sears, W. Timp, Epigenetic changes [130] G.L. Simon, M.S. Klemper, D.L. Kasper, S.L. Gorbach,
induced by Bacteroides fragilis toxin (BFT), Infect. Immun. Alterations in opsonophagocytic killing by neutrophils
(2019), https://doi.org/10.1128/IAI.00447-18. of Bacteroides fragilis associated with animal and labora-
[119] S. Rabizadeh, K.-J. Rhee, S. Wu, D. Huso, C.M. Gan, tory passage: effect of capsular polysaccharide, J. Infect.
J.E. Golub, X. Wu, M. Zhang, C.L. Sears, Enterotoxigenic Dis. 145 (1982) 72e77, https://doi.org/10.1093/infdis/
bacteroides fragilis: a potential instigator of colitis, Inflamm. 145.1.72.
Bowel Dis. 13 (2007) 1475e1483, https://doi.org/10.1002/ [131] A.O. Tzianabos, A.B. Onderdonk, B. Rosner, R.L. Cisneros,
ibd.20265. D.L. Kasper, Structural features of polysaccharides that
[120] K.-J. Rhee, S. Wu, X. Wu, D.L. Huso, B. Karim, induce intra-abdominal abscesses, Science 262 (1993)
A.A. Franco, S. Rabizadeh, J.E. Golub, L.E. Mathews, 416e419.
J. Shin, R.B. Sartor, D. Golenbock, A.R. Hamad, C.M. Gan, [132] Y. Wang, W.M. Kalka-Moll, M.H. Roehrl, D.L. Kasper,
F. Housseau, C.L. Sears, Induction of persistent colitis by a Structural basis of the abscess-modulating polysaccharide
human commensal, enterotoxigenic Bacteroides fragilis, in A2 from Bacteroides fragilis, Proc. Natl. Acad. Sci. U. S. A
Good gone bad 781

97 (2000) 13478e13483, https://doi.org/10.1073/ olytic human gut bacterial symbiont, Cell Host Microbe 4
pnas.97.25.13478. ( 2 0 0 8 ) 4 4 7 e4 5 7 , h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 /
[133] C.M. Krinos, M.J. Coyne, K.G. Weinacht, A.O. Tzianabos, j.chom.2008.09.007.
D.L. Kasper, L.E. Comstock, Extensive surface diversity of [146] A. Marcobal, M. Barboza, E.D. Sonnenburg, N. Pudlo,
a commensal microorganism by multiple DNA inversions, E.C. Martens, P. Desai, C.B. Lebrilla, B.C. Weimer,
Nature 414 (2001) 555e558, https://doi.org/10.1038/ D.A. Mills, J.B. German, J.L. Sonnenburg, Bacteroides in
35107092. the infant gut consume milk oligosaccharides via mucus-
[134] M.J. Coyne, A.O. Tzianabos, B.C. Mallory, V.J. Carey, utilization pathways, Cell Host Microbe 10 (2011) 507e514,
D.L. Kasper, L.E. Comstock, Polysaccharide biosynthesis https://doi.org/10.1016/j.chom.2011.10.007.
locus required for virulence of Bacteroides fragilis, Infect. [147] Y. Cao, E.R. Rocha, C.J. Smith, Efficient utilization of
Immun. 69 (2001) 4342e4350, https://doi.org/10.1128/ complex N-linked glycans is a selective advantage for
IAI.69.7.4342-4350.2001. <em>Bacteroides fragilis</em> in extraintestinal infec-
[135] M.J. Coyne, M. Chatzidaki-Livanis, L.C. Paoletti, tions, Proc. Natl. Acad. Sci. 111 (2014) 12901, https://
L.E. Comstock, Role of glycan synthesis in colonization doi.org/10.1073/pnas.1407344111. LP e 12906.
of the mammalian gut by the bacterial symbiont Bacter- [148] G.P. Donaldson, M.S. Ladinsky, K.B. Yu, J.G. Sanders,
oides fragilis, Proc. Natl. Acad. Sci. U. S. A 105 (2008) B.B. Yoo, W.-C. Chou, M.E. Conner, A.M. Earl, R. Knight,
13099e13104, https://doi.org/10.1073/pnas.0804220105. P.J. Bjorkman, S.K. Mazmanian, Gut microbiota utilize
[136] C.H. Liu, S.M. Lee, J.M. Vanlare, D.L. Kasper, immunoglobulin A for mucosal colonization, Science 360
S.K. Mazmanian, Regulation of surface architecture by (2018) 795e800, https://doi.org/10.1126/science.aaq0926.
symbiotic bacteria mediates host colonization, Proc. Natl. [149] M.R. Popoff, Clostridial pore-forming toxins: powerful
Acad. Sci. U. S. A 105 (2008) 3951e3956, https://doi.org/ virulence factors, Anaerobe 30 (2014) 220e238, https://
10.1073/pnas.0709266105. doi.org/10.1016/j.anaerobe.2014.05.014.
[137] B.A. Cobb, Q. Wang, A.O. Tzianabos, D.L. Kasper, [150] B.J. Berube, J. Bubeck Wardenburg, Staphylococcus
Polysaccharide processing and presentation by the MHCII aureus alpha-toxin: nearly a century of intrigue, Toxins 5
pathway, Cell 117 (2004) 677e687, https://doi.org/ (2013) 1140e1166.
10.1016/J.CELL.2004.05.001. [151] M. Bielaszewska, T. Aldick, A. Bauwens, H. Karch,
[138] B.A. Cobb, D.L. Kasper, Characteristics of carbohydrate Hemolysin of enterohemorrhagic Escherichia coli: struc-
antigen binding to the presentation protein HLA-DR, ture, transport, biological activity and putative role in
Glycobiology 18 (2008) 707e718, https://doi.org/10.1093/ virulence, Int. J. Med. Microbiol. 304 (2014) 521e529,
glycob/cwn050. https://doi.org/10.1016/j.ijmm.2014.05.005.
[139] Yong-Hoon Choi, Michael H. Roehrl, Dennis L. Kasper, [152] K.P. Robertson, C.J. Smith, A.M. Gough, E.R. Rocha,
Julia Y. Wang, A Unique Structural Pattern Shared by T- Characterization of Bacteroides fragilis hemolysins and
Cell-Activating and Abscess-Regulating Zwitterionic Poly- regulation and synergistic interactions of HlyA and HlyB,
saccharidesy, 2002, https://doi.org/10.1021/BI020491V. Infect. Immun. 74 (2006) 2304e2316, https://doi.org/
[140] L.E. Blandford, E.L. Johnston, J.D. Sanderson, W.G. Wade, 10.1128/IAI.74.4.2304-2316.2006.
A.J. Lax, Promoter orientation of the immunomodulatory [153] L.A. Lobo, A.L. Jenkins, C. Jeffrey Smith, E.R. Rocha,
Bacteroides fragilis capsular polysaccharide A (PSA) is off in Expression of Bacteroides fragilis hemolysins in vivo and role
individuals with inflammatory bowel disease (IBD), Gut of HlyBA in an intra-abdominal infection model, Microbiology 2
M i c r o b . ( 2 0 1 9 ) 1 e9 , h t t p s : / / d o i . o r g / 1 0 . 1 0 8 0 / (2013) 326e337, https://doi.org/10.1002/mbo3.76.
19490976.2018.1560755. [154] A.S. Luis, E.C. Martens, Interrogating gut bacterial gen-
[141] J.L. Chan, S. Wu, A.L. Geis, G. V Chan, T.A.M. Gomes, omes for discovery of novel carbohydrate degrading
S.E. Beck, X. Wu, H. Fan, A.J. Tam, L. Chung, H. Ding, enzymes, Curr. Opin. Chem. Biol. 47 (2018) 126e133,
H. Wang, D.M. Pardoll, F. Housseau, C.L. Sears, Non- https://doi.org/10.1016/j.cbpa.2018.09.012.
toxigenic Bacteroides fragilis (NTBF) administration reduces [155] A.G. FRASER, R. BROWN, Neuraminidase production by
bacteria-driven chronic colitis and tumor development inde- bacteroidaceae, J. Med. Microbiol. 14 (1981) 63e76,
pendent of polysaccharide A, Mucosal Immunol. 12 (2019) https://doi.org/10.1099/00222615-14-1-63.
164e177, https://doi.org/10.1038/s41385-018-0085-5. [156] N. Juge, L. Tailford, C.D. Owen, Sialidases from gut
[142] S.M. Lee, G.P. Donaldson, Z. Mikulski, S. Boyajian, K. Ley, bacteria: a mini-review, Biochem. Soc. Trans. 44 (2016)
S.K. Mazmanian, Bacterial colonization factors control 166e175, https://doi.org/10.1042/BST20150226.
specificity and stability of the gut microbiota, Nature 501 [157] B.L. Haines-Menges, W.B. Whitaker, J.B. Lubin, E.F. Boyd,
(2013) 426e429, https://doi.org/10.1038/nature12447. Host sialic acids: a delicacy for the pathogen with
[143] E.C. Martens, E.C. Lowe, H. Chiang, N.A. Pudlo, M. Wu, discerning taste, Microbiol. Spectr. 3 (2015), https://
N.P. McNulty, D.W. Abbott, B. Henrissat, H.J. Gilbert, doi.org/10.1128/microbiolspec.MBP-0005-2014.
D.N. Bolam, J.I. Gordon, Recognition and degradation of [158] V.G. Godoy, M.M. Dallas, T.A. Russo, M.H. Malamy, A role
plant cell wall polysaccharides by two human gut sym- for Bacteroides fragilis neuraminidase in bacterial growth in
bionts, PLoS Biol. 9 (2011), e1001221, https://doi.org/ two model systems, Infect. Immun. 61 (1993) 4415e4426.
10.1371/journal.pbio.1001221. http://www.ncbi.nlm.nih.gov/pubmed/8406832. (Accessed
[144] J.L. Sonnenburg, J. Xu, D.D. Leip, C.-H. Chen, 31 July 2019).
B.P. Westover, J. Weatherford, J.D. Buhler, J.I. Gordon, [159] R.M.C.P. Domingues, S.M.B. Cavalcanti, A.F.B. Andrade,
Glycan foraging in vivo by an intestine-adapted bacterial M.C.S. Ferreira, Sialic acid as receptor of Bacteroides
symbiont, Science 307 (2005) 1955e1959, https://doi.org/ fragilis lectin-like adhesin, Zentralblatt Bakteriol. 277 (1992)
10.1126/science.1109051. 340e344, https://doi.org/10.1016/S0934-8840(11)80912-6.
[145] E.C. Martens, H.C. Chiang, J.I. Gordon, Mucosal glycan [160] C.A. Guzma n, M. Plate
, C. Pruzzo, M.J. Gauthier, Role of
foraging enhances fitness and transmission of a sacchar- neuraminidase-dependent adherence in Bacteroides fragi-
782 Good gone bad

lis attachment to human epithelial cells, FEMS Microbiol. [174] H. Pauer, E. de O. Ferreira, J. dos Santos-Filho,
Lett. 59 (1990) 187e192, https://doi.org/10.1111/j.1574- M.B. Portela, R.B. Zingali, R.M.A. Soares,
6968.1989.tb03107.x. R.M.C.P. Domingues, A TonB-dependent outer membrane
[161] R. Jotwani, N. Kato, H. Kato, K. Watanabe, K. Ueno, protein as a Bacteroides fragilis fibronectin-binding mole-
Detection of Bacteroides fragilis in clinical specimens by cule, FEMS Immunol. Med. Microbiol. 55 (2009) 388e395,
polymerase chain reaction amplification of the neuramini- https://doi.org/10.1111/j.1574-695X.2009.00532.x.
dase gene, Curr. Microbiol. 31 (1995) 215e219, https:// [175] H. Pauer, S.N.V. Cavalcanti, F.L. Teixeira, J. Santos-Filho,
doi.org/10.1007/BF00298376. R.C. Vommaro, A.C.S.C. Oliveira, E.O. Ferreira,
[162] R.F. Thornton, T.F. Kagawa, P.W. O'Toole, J.C. Cooney, The R.R.M.C.P. Domingues, Inactivation of a fibronectin-bind-
dissemination of C10 cysteine protease genes in Bacteroides ing TonB-dependent protein increases adhesion properties
fragilis by mobile genetic elements, BMC Microbiol. 10 (2010) of Bacteroides fragilis, J. Med. Microbiol. 62 (2013)
122, https://doi.org/10.1186/1471-2180-10-122. 1524e1530, https://doi.org/10.1099/jmm.0.054692-0.
[163] R.F. Thornton, E.C. Murphy, T.F. Kagawa, P.W. O'Toole, [176] P. Eiring, B. Manncke, K. Gerbracht, H. Werner, Bacter-
J.C. Cooney, The effect of environmental conditions on oides fragilis adheres to laminin significantly stronger than
expression of Bacteroides fragilis and Bacteroides thetaio- bacteroides thetaiotaomicron and other species of the
taomicron C10 protease genes, BMC Microbiol. 12 (2012) genus, Zentralblatt Bakteriol. 282 (1995) 279e286, https://
190, https://doi.org/10.1186/1471-2180-12-190. doi.org/10.1016/S0934-8840(11)80128-3.
[164] Y. Chen, T. Kinouchi, K. Kataoka, S. Akimoto, Y. Ohnishi, [177] E. de O. Ferreira, L. Araújo Lobo, D. Barreiros Petro polis,
Purification and characterization of a fibrinogen-degrading K.E. dos, S. Avelar, M.C. Ferreira, F.C. e Silva Filho,
protease in Bacteroides fragilis strain YCH46, Microbiol. R.M.C.P. Domingues, A Bacteroides fragilis surface
Immunol. 39 (1995) 967e977, https://doi.org/10.1111/ glycoprotein mediates the interaction between the bacter-
j.1348-0421.1995.tb03300.x. ium and the extracellular matrix component laminin-1, Res.
[165] S. Houston, G.W. Blakely, A. McDowell, L. Martin, Microbiol. 157 (2006) 960e966, https://doi.org/10.1016/
S. Patrick, Binding and degradation of fibrinogen by J.RESMIC.2006.09.005.
Bacteroides fragilis and characterization of a 54 kDa [178] E. de O. Ferreira, F.L. Teixeira, F. Cordeiro, L. Araujo Lobo,
fibrinogen-binding protein, Microbiology 156 (2010) E.R. Rocha, J.C. Smith, R.M.C.P. Domingues, The Bfp60
2516e2526, https://doi.org/10.1099/mic.0.038588-0. surface adhesin is an extracellular matrix and plasminogen
[166] P. Klemm, M.A. Schembri, Bacterial adhesins: function and protein interacting in Bacteroides fragilis, Int. J. Med.
structure, Int. J. Med. Microbiol. 290 (2000) 27e35, https:// Microbiol. 303 (2013) 492e497, https://doi.org/10.1016/
doi.org/10.1016/S1438-4221(00)80102-2. J.IJMM.2013.06.007.
[167] K. Moonens, H. Remaut, Evolution and structural dynamics [179] E. de O. Ferreira, J.B. de Carvalho, R.J.M. Peixoto,
of bacterial glycan binding adhesins, Curr. Opin. Struct. L.A. Lobo, R.B. Zingalli, C.J. Smith, E.R. Rocha,
Biol. 44 (2017) 48e58, https://doi.org/10.1016/ R.M.C.P. Domingues, The interaction of Bacteroides
j.sbi.2016.12.003. fragilis with components of the human fibrinolytic system,
[168] M.K. Hospenthal, T.R.D. Costa, G. Waksman, FEMS Immunol. Med. Microbiol. 56 (2009) 48e55, https://
A comprehensive guide to pilus biogenesis in Gram- doi.org/10.1111/j.1574-695X.2009.00546.x.
negative bacteria, Nat. Rev. Microbiol. 15 (2017) [180] E.C. Murphy, M. Morgelin, J.C. Cooney, I.-M. Frick, Interaction
365e379, https://doi.org/10.1038/nrmicro.2017.40. of Bacteroides fragilis and Bacteroides thetaiotaomicron with
[169] I. Brook, L.A. Myhal, C.H. Dorsey, Encapsulation and pilus the kallikrein-kinin system, Microbiology 157 (2011)
formation of Bacteroides spp. in normal flora abscesses 2094e2105, https://doi.org/10.1099/mic.0.046862-0.
and blood, J. Infect. (1992), https://doi.org/10.1016/0163- [181] R. Sijbrandi, M. Stork, J. Luirink, B.R. Otto, Pbp, a cell-
4453(92)91479-U. surface exposed plasminogen binding protein of Bacter-
[170] C. Pruzzo, B. Dainelli, M. Ricchetti, Piliated Bacteroides oides fragilis, Microb. Infect. 10 (2008) 514e521, https://
fragilis strains adhere to epithelial cells and are more doi.org/10.1016/J.MICINF.2008.01.015.
sensitive to phagocytosis by human neutrophils than [182] E.S. Friedman, K. Bittinger, T. V Esipova, L. Hou, L. Chau,
nonpiliated strains, Infect. Immun. 43 (1984) 189e194. J. Jiang, C. Mesaros, P.J. Lund, X. Liang, G.A. FitzGerald,
http://www.ncbi.nlm.nih.gov/pubmed/6140223. (Accessed M. Goulian, D. Lee, B.A. Garcia, I.A. Blair, S.A. Vinogradov,
31 July 2019). G.D. Wu, Microbes vs. chemistry in the origin of the
[171] W.A.C. Vel, F. Namavar, A.M.J.J. Verweij-Van Vught, anaerobic gut lumen, Proc. Natl. Acad. Sci. U. S. A 115
A.N.B. Pubben, D.M. Maclaren, Haemagglutination by the ( 2 0 1 8 ) 4 1 7 0 e4 1 7 5 , h t t p s : / / d o i . o r g / 1 0 . 1 0 7 3 /
Bacteroides fragilis group, J. Med. Microbiol. 21 (1986) pnas.1718635115.
105e107, https://doi.org/10.1099/00222615-21-2-105. [183] R.D. Rolfe, D.J. Hentges, J.T. Barrett, B.J. Campbell,
[172] V. Nakano, M.J. Avila-Campos, Virulence markers and Oxygen tolerance of human intestinal anaerobes, Am. J.
antimicrobial susceptibility of bacteria of the Bacteroides Clin. Nutr. 30 (1977) 1762e1769, https://doi.org/10.1093/
fragilis group isolated from stool of children with diarrhea ajcn/30.11.1762.
in Sa ~o Paulo, Brazil, Mem. Inst. Oswaldo Cruz 99 (2004) [184] E. Rocha, C.D. Herren, D.J. Smalley, C. Smith, The
3 0 7 e3 1 2 , h t t p s : / / d o i . o r g / 1 0 . 1 5 9 0 / S 0 0 7 4 - complex oxidative stress response of Bacteroides fragilis:
02762004000300012. the role of OxyR in control of gene expression, Anaerobe 9
[173] C. Pruzzo, C.A. Guzma n, B. Dainelli, Incidence of (2003) 165e173, https://doi.org/10.1016/S1075-9964(03)
hemagglutination activity among pathogenic and non- 00118-5.
pathogenic Bacteroides fragilis strains and role of capsule [185] C.D. Herren, E.R. Rocha, C.J. Smith, Genetic analysis of
and pili in HA and adherence, FEMS Microbiol. Lett. 59 an important oxidative stress locus in the anaerobe
(1989) 113e118, https://doi.org/10.1016/0378-1097(89) Bacteroides fragilis, Gene 316 (2003) 167e175, https://
90469-2. doi.org/10.1016/S0378-1119(03)00759-5.
Good gone bad 783

[186] C.J. Sund, E.R. Rocha, A.O. Tzinabos, W.G. Wells, [199] F.L. Teixeira, D.N. Silva, H. Pauer, L.Q. Ferreira, O.
J.M. Gee, M.A. Reott, D.P. O'Rourke, C.J. Smith, The Ferreira E. de, R.M. Domingues, L.A. Lobo, The role of
Bacteroides fragilis transcriptome response to oxygen and BmoR, a MarR Family Regulator, in the survival of
H2O2: the role of OxyR and its effect on survival and Bacteroides fragilis during oxidative stress, Int. J. Med.
virulence, Mol. Microbiol. 67 (2007) 129e142, https:// Microbiol. 303 (2013) 443e448, https://doi.org/10.1016/
doi.org/10.1111/j.1365-2958.2007.06031.x. J.IJMM.2013.05.007.
[187] E. de Oliveira Ferreira, E. Yates, M. Jourlin, J. Liu, [200] F.L. Teixeira, H. Pauer, S.B. Costa, C.J. Smith,
R.M.C. Pilotto Domingues, M. Goldner, Anaerobe/aerobe R.M.C.P. Domingues, E.R. Rocha, L.A. Lobo, Deletion of
environmental flux determines protein expression profiles BmoR affects the expression of genes related to thiol/
of Bacteroides fragilis, a redox pathogen, Anaerobe 17 disulfide balance in Bacteroides fragilis, Sci. Rep. 8 (2018)
( 2 0 1 1 ) 4 e1 4 , h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 / J . A N A E - 14405, https://doi.org/10.1038/s41598-018-32880-7.
ROBE.2010.09.004. [201] L. Pumbwe, C.A. Skilbeck, V. Nakano, M.J. Avila-Campos,
[188] A.D. Baughn, M.H. Malamy, The strict anaerobe Bacter- R.M.F. Piazza, H.M. Wexler, Bile salts enhance bacterial
oides fragilis grows in and benefits from nanomolar co-aggregation, bacterial-intestinal epithelial cell adhesion,
concentrations of oxygen, Nature 427 (2004) 441e444, biofilm formation and antimicrobial resistance of Bacter-
https://doi.org/10.1038/nature02285. oides fragilis, Microb. Pathog. 43 (2007) 78e87, https://
[189] B.M. Meehan, A.D. Baughn, R. Gallegos, M.H. Malamy, doi.org/10.1016/J.MICPATH.2007.04.002.
Inactivation of a single gene enables microaerobic growth [202] R.F. Boente, H. Pauer, D.N.S. Silva, J.S. Filho, V. Sandim,
of the obligate anaerobe Bacteroides fragilis, Proc. Natl. L.C.M. Antunes, R.B.R. Ferreira, R.B. Zingali,
Acad. Sci. 109 (2012) 12153e12158, https://doi.org/ R.M.C.P. Domingues, L.A. Lobo, Differential proteomic
10.1073/pnas.1203796109. analysis of outer membrane enriched extracts of Bacter-
[190] F.P. Tally, B.R. Goldin, N. V Jacobus, S.L. Gorbach, oides fragilis grown under bile salts stress, Anaerobe 39
Superoxide dismutase in anaerobic bacteria of clinical ( 2 0 1 6 ) 8 4 e9 0 , h t t p s : / / d o i . o r g / 1 0 . 1 0 1 6 / J . A N A E -
significance, Infect. Immun. 16 (1977) 20e25. http://www. ROBE.2016.03.003.
ncbi.nlm.nih.gov/pubmed/326669. (Accessed 31 July [203] C.M.G. Silva, D.N. dos S Silva, S.B. da Costa, J.S. de
2019). S. Almeida, R.F. Boente, F.L. Teixeira,
[191] E.R. Rocha, T. Selby, J.P. Coleman, C.J. Smith, Oxidative R.M.C.P. Domingues, L.A. Lobo, Inactivation of MarR gene
stress response in an anaerobe, Bacteroides fragilis: a role homologs increases susceptibility to antimicrobials in
for catalase in protection against hydrogen peroxide, Bacteroides fragilis, Braz. J. Microbiol. 49 (2018)
J. Bacteriol. 178 (1996) 6895e6903, https://doi.org/ 200e206, https://doi.org/10.1016/J.BJM.2017.05.005.
10.1128/jb.178.23.6895-6903.1996. [204] A. Konovalova, D.E. Kahne, T.J. Silhavy, Outer membrane
[192] E.R. Rocha, C.J. Smith, Role of the alkyl hydroperoxide biogenesis, Annu. Rev. Microbiol. 71 (2017) 539e556,
reductase (ahpCF) gene in oxidative stress defense of the https://doi.org/10.1146/annurev-micro-090816-093754.
obligate Anaerobe bacteroides fragilis, J. Bacteriol. 181 [205] P. Sperandeo, A.M. Martorana, A. Polissi, Lipopolysac-
(1999) 5701e5710. http://www.ncbi.nlm.nih.gov/pubmed/ charide biosynthesis and transport to the outer membrane
10482511. (Accessed 31 July 2019). of gram-negative bacteria, Subcell. Biochem. (2019) 9e37,
[193] E.R. Rocha, S.C. Andrews, J.N. Keen, J.H. Brock, Isolation https://doi.org/10.1007/978-3-030-18768-2_2.
of a ferritin from Bacteroides fragilis, FEMS Microbiol. Lett. [206] B.W. Simpson, M.S. Trent, Pushing the envelope: LPS
95 (1992) 207e212, https://doi.org/10.1111/j.1574- modifications and their consequences, Nat. Rev. Microbiol.
6968.1992.tb05367.x. 17 (2019) 403e416, https://doi.org/10.1038/s41579-019-
[194] E.R. Rocha, C.J. Smith, Transcriptional regulation of the 0201-x.
Bacteroides fragilis ferritin gene (ftnA) by redox stress, [207] Y.-C. Lu, W.-C. Yeh, P.S. Ohashi, LPS/TLR4 signal
Microbiology 150 (2004) 2125e2134, https://doi.org/ transduction pathway, Cytokine 42 (2008) 145e151,
10.1099/mic.0.26948-0. https://doi.org/10.1016/j.cyto.2008.01.006.
[195] G.H. Gauss, M.A. Reott, E.R. Rocha, M.J. Young, [208] M.J. Wannemuehler, S.M. Michalek, E. Jirillo,
T. Douglas, C.J. Smith, C.M. Lawrence, Characterization S.I. Williamson, M. Hirasawa, J.R. McGhee, LPS regula-
of the Bacteroides fragilis bfr gene product identifies a tion of the immune response: Bacteroides endotoxin
bacterial DPS-like protein and suggests evolutionary links induces mitogenic, polyclonal, and antibody responses in
in the ferritin superfamily, J. Bacteriol. 194 (2012) 15e27, classical LPS responsive but not C3H/HeJ mice,
https://doi.org/10.1128/JB.05260-11. J. Immunol. 133 (1984) 299e305. http://www.ncbi.nlm.
[196] M.I. Betteken, E.R. Rocha, C.J. Smith, Dps and DpsL nih.gov/pubmed/6202784. (Accessed 31 July 2019).
mediate survival in vitro and in vivo during the prolonged [209] D.L. Kasper, Chemical and biological characterization of
oxidative stress response in Bacteroides fragilis, the lipopolysaccharide of Bacteroides fragilis subspecies
J. Bacteriol. 197 (2015) 3329e3338, https://doi.org/ fragilis, J. Infect. Dis. 134 (1976) 59e66, https://doi.org/
10.1128/JB.00342-15. 10.1093/infdis/134.1.59.
[197] M.A. Reott, A.C. Parker, E.R. Rocha, C.J. Smith, Thior- [210] A.A. Lindberg, A. Weintraub, U. Za €hringer, E. Th Rietschel,
edoxins in redox maintenance and survival during oxidative Structure-Activity Relationships in Lipopolysaccharides
stress of Bacteroides fragilis, J. Bacteriol. 191 (2009) of Bacteroides Fragilis, n.d. https://www.jstor.org/stable/
3384e3391, https://doi.org/10.1128/JB.01665-08. pdf/4455470.pdf?refreqid¼excelsior%3A44d8641961de40a
[198] E.R. Rocha, G. Owens, C.J. Smith, The redox-sensitive a8dd9fbbe16b7be53 (accessed July 31, 2019)..
transcriptional activator OxyR regulates the peroxide [211] B. Brake, C. Larcher, T.F. Schulz, W. Prodinger,
response regulon in the obligate anaerobe Bacteroides M.P. Dierich, Species specific monoclonal antibodies
fragilis, J. Bacteriol. 182 (2000) 5059e5069, https://doi.org/ to Bacteroides fragilis lipopolysaccharide protect
10.1128/jb.182.18.5059-5069.2000. mice from severe infection, Zentralblatt Bakteriol. 277
784 Good gone bad

(1992) 320e328, https://doi.org/10.1016/S0934-8840(11) T. Vatanen, C. Huttenhower, R.J. Xavier, M. Rewers,


80909-6. W. Hagopian, J. Toppari, A.-G. Ziegler, J.-X. She,
[212] H. Seedorf, N.W. Griffin, V.K. Ridaura, A. Reyes, J. Cheng, B. Akolkar, A. Lernmark, H. Hyoty, K. Vehik,
F.E. Rey, M.I. Smith, G.M. Simon, R.H. Scheffrahn, J.P. Krischer, J.F. Petrosino, Temporal development of
D. Woebken, A.M. Spormann, W. Van Treuren, the gut microbiome in early childhood from the TEDDY
L.K. Ursell, M. Pirrung, A. Robbins-Pianka, B.L. Cantarel, study, Nature 562 (2018) 583e588, https://doi.org/10.1038/
V. Lombard, B. Henrissat, R. Knight, J.I. Gordon, Bacteria s41586-018-0617-x.
from diverse habitats colonize and compete in the mouse [226] K. Korpela, W.M. de Vos, Early life colonization of the
gut, Cell 159 (2014) 253e266, https://doi.org/10.1016/ human gut: microbes matter everywhere, Curr. Opin.
j.cell.2014.09.008. Microbiol. 44 (2018) 70e78, https://doi.org/10.1016/
[213] M.A. Bauer, K. Kainz, D. Carmona-Gutierrez, F. Madeo, j.mib.2018.06.003.
Microbial wars: competition in ecological niches and within [227] H. Renz, P. Brandtzaeg, M. Hornef, The impact of perinatal
the microbiome, Microb. Cell (Graz, Austria) 5 (2018) immune development on mucosal homeostasis and
215e219, https://doi.org/10.15698/mic2018.05.628. chronic inflammation, Nat. Rev. Immunol. 12 (2011)
[214] M.E. Hibbing, C. Fuqua, M.R. Parsek, S.B. Peterson, 9e23, https://doi.org/10.1038/nri3112.
Bacterial competition: surviving and thriving in the microbial [228] S. Rokhsefat, A. Lin, E.M. Comelli, Mucin-microbiota
jungle, Nat. Rev. Microbiol. 8 (2010) 15e25, https://doi.org/ interaction during postnatal maturation of the intestinal
10.1038/nrmicro2259. ecosystem: clinical implications, Dig. Dis. Sci. 61 (2016)
[215] A. Jamet, X. Nassif, New players in the toxin field: 1473e1486, https://doi.org/10.1007/s10620-016-4032-6.
polymorphic toxin systems in bacteria, mBio 6 (2015), [229] L. Arike, G.C. Hansson, The densely O-glycosylated MUC2
https://doi.org/10.1128/mBio.00285-15 e00285-15. mucin protects the intestine and provides food for the
[216] E.C. Garcia, Contact-dependent interbacterial toxins deli- commensal bacteria, J. Mol. Biol. 428 (2016) 3221e3229,
ver a message, Curr. Opin. Microbiol. 42 (2018) 40e46, https://doi.org/10.1016/j.jmb.2016.02.010.
https://doi.org/10.1016/j.mib.2017.09.011. [230] M.E. V Johansson, J.M.H. Larsson, G.C. Hansson, The two
[217] T.G. Dong, B.T. Ho, D.R. Yoder-Himes, J.J. Mekalanos, mucus layers of colon are organized by the MUC2 mucin,
Identification of T6SS-dependent effector and immunity whereas the outer layer is a legislator of hostemicrobial
proteins by Tn-seq in Vibrio cholerae, Proc. Natl. Acad. Sci. interactions, Proc. Natl. Acad. Sci. 108 (2011) 4659, https://
U. S. A 110 (2013) 2623e2628, https://doi.org/10.1073/ doi.org/10.1073/pnas.1006451107. LP e 4665.
pnas.1222783110. [231] T. Pelaseyed, J.H. Bergstrom, J.K. Gustafsson, A. Ermund,
[218] T.G. Sana, N. Flaugnatti, K.A. Lugo, L.H. Lam, A. Jacobson, G.M.H. Birchenough, A. Schutte, S. van der Post,
V. Baylot, E. Durand, L. Journet, E. Cascales, D.M. Monack, F. Svensson, A.M. Rodriguez-Pineiro, E.E.L. Nystrom,
Salmonella Typhimurium utilizes a T6SS-mediated antibac- C. Wising, M.E. V Johansson, G.C. Hansson, The mucus
terial weapon to establish in the host gut, Proc. Natl. Acad. Sci. and mucins of the goblet cells and enterocytes provide the
U. S. A 113 (2016) E5044eE5051, https://doi.org/10.1073/ first defense line of the gastrointestinal tract and interact
pnas.1608858113. with the immune system, Immunol. Rev. 260 (2014) 8e20,
[219] S.L. Logan, J. Thomas, J. Yan, R.P. Baker, D.S. Shields, https://doi.org/10.1111/imr.12182.
J.B. Xavier, B.K. Hammer, R. Parthasarathy, The Vibrio [232] A.M. Rodríguez-Pin ~ eiro, M.E. V Johansson, The colonic
cholerae type VI secretion system can modulate host mucus protection depends on the microbiota, Gut Microb. 6
intestinal mechanics to displace gut bacterial symbionts, ( 2 0 1 5 ) 3 2 6 e3 3 0 , h t t p s : / / d o i . o r g / 1 0 . 1 0 8 0 /
Proc. Natl. Acad. Sci. U. S. A 115 (2018) E3779eE3787, 19490976.2015.1086057.
https://doi.org/10.1073/pnas.1720133115. [233] H.E. Jakobsson, A.M. Rodríguez-Pin ~ eiro, A. Schütte,
[220] M.J. Coyne, L.E. Comstock, Type VI secretion systems and A. Ermund, P. Boysen, M. Bemark, F. Sommer,
the gut microbiota, Microbiol. Spectr. 7 (2019), https:// F. Ba €ckhed, G.C. Hansson, M.E. V Johansson, The
doi.org/10.1128/microbiolspec.PSIB-0009-2018. composition of the gut microbiota shapes the colon mucus
[221] Y. Cherrak, C. Rapisarda, R. Pellarin, G. Bouvier, barrier, EMBO Rep. 16 (2015) 164e177, https://doi.org/
B. Bardiaux, F. Allain, C. Malosse, M. Rey, J. Chamot- 10.15252/embr.201439263.
Rooke, E. Cascales, R. Fronzes, E. Durand, Biogenesis [234] L. Aymeric, F. Donnadieu, C. Mulet, L. du Merle, G. Nigro,
and structure of a type VI secretion baseplate, Nat. A. Saffarian, M. Berard, C. Poyart, S. Robine, B. Regnault,
Microbiol. 3 (2018) 1404e1416, https://doi.org/10.1038/ P. Trieu-Cuot, P.J. Sansonetti, S. Dramsi, Colorectal
s41564-018-0260-1. cancer specific conditions promote Streptococcus galloly-
[222] C.J. Alteri, H.L.T. Mobley, The versatile type VI secretion ticus gut colonization, Proc. Natl. Acad. Sci. U. S. A 115
system, Microbiol. Spectr. 4 (2016), https://doi.org/10.1128/ (2018) E283eE291, https://doi.org/10.1073/pnas.
microbiolspec.VMBF-0026-2015. 1715112115.
[223] A. Hachani, T.E. Wood, A. Filloux, Type VI secretion and [235] K.W. Kinzler, B. Vogelstein, Lessons from hereditary
anti-host effectors, Curr. Opin. Microbiol. 29 (2016) 81e93, colorectal cancer, Cell 87 (1996) 159e170.
https://doi.org/10.1016/j.mib.2015.11.006. [236] A.J. Rowan, H. Lamlum, M. Ilyas, J. Wheeler, J. Straub,
[224] B.D. Ross, A.J. Verster, M.C. Radey, D.T. Schmidtke, A. Papadopoulou, D. Bicknell, W.F. Bodmer, I.P. Tomlinson,
C.E. Pope, L.R. Hoffman, A.M. Hajjar, S.B. Peterson, APC mutations in sporadic colorectal tumors: a mutational
E. Borenstein, J.D. Mougous, Human gut bacteria contain “hotspot” and interdependence of the “two hits”, Proc. Natl.
acquired interbacterial defence systems, Nature 575 (2019) Acad. Sci. U. S. A 97 (2000) 3352e3357.
224e228, https://doi.org/10.1038/s41586-019-1708-z. [237] L. Zhang, J.W. Shay, Multiple roles of APC and its
[225] C.J. Stewart, N.J. Ajami, J.L. O'Brien, D.S. Hutchinson, therapeutic implications in colorectal cancer, J. Natl.
D.P. Smith, M.C. Wong, M.C. Ross, R.E. Lloyd, Cancer Inst. 109 (2017), https://doi.org/10.1093/jnci/
H. Doddapaneni, G.A. Metcalf, D. Muzny, R.A. Gibbs, djw332.
Good gone bad 785

[238] A.R. Moser, E.M. Mattes, W.F. Dove, M.J. Lindstrom, H.M. O'Hagan, C.L. Sears, Reduction of murine colon
J.D. Haag, M.N. Gould, ApcMin, a mutation in the murine tumorigenesis driven by enterotoxigenic Bacteroides
Apc gene, predisposes to mammary carcinomas and focal fragilis using cefoxitin treatment, J. Infect. Dis. 214
alveolar hyperplasias, Proc. Natl. Acad. Sci. U. S. A 90 (2016) 122e129, https://doi.org/10.1093/infdis/jiw069.
(1993) 8977e8981. [245] N. Zmora, G. Zilberman-Schapira, J. Suez, U. Mor, M. Dori-
[239] S. Wu, K.-J. Rhee, E. Albesiano, S. Rabizadeh, X. Wu, H.- Bachash, S. Bashiardes, E. Kotler, M. Zur, D. Regev-
R. Yen, D.L. Huso, F.L. Brancati, E. Wick, F. McAllister, Lehavi, R.B.-Z. Brik, S. Federici, Y. Cohen, R. Linevsky,
F. Housseau, D.M. Pardoll, C.L. Sears, A human colonic D. Rothschild, A.E. Moor, S. Ben-Moshe, A. Harmelin,
commensal promotes colon tumorigenesis via activation of S. Itzkovitz, N. Maharshak, O. Shibolet, H. Shapiro,
T helper type 17 T cell responses, Nat. Med. 15 (2009) M. Pevsner-Fischer, I. Sharon, Z. Halpern, E. Segal,
1016e1022, https://doi.org/10.1038/nm.2015. E. Elinav, Personalized gut mucosal colonization resis-
[240] S. Tomkovich, C.M. Dejea, K. Winglee, J.L. Drewes, tance to empiric probiotics is associated with unique host
L. Chung, F. Housseau, J.L. Pope, J. Gauthier, X. Sun, and microbiome features, Cell 174 (2018) 1388e1405,
M. Muhlbauer, X. Liu, P. Fathi, R.A. Anders, S. Besharati, https://doi.org/10.1016/j.cell.2018.08.041, e21.
E. Perez-Chanona, Y. Yang, H. Ding, X. Wu, S. Wu, [246] J. Suez, N. Zmora, G. Zilberman-Schapira, U. Mor, M. Dori-
J.R. White, R.Z. Gharaibeh, A.A. Fodor, H. Wang, Bachash, S. Bashiardes, M. Zur, D. Regev-Lehavi, R. Ben-
D.M. Pardoll, C. Jobin, C.L. Sears, Human colon mucosal Zeev Brik, S. Federici, M. Horn, Y. Cohen, A.E. Moor,
biofilms from healthy or colon cancer hosts are carcino- D. Zeevi, T. Korem, E. Kotler, A. Harmelin, S. Itzkovitz,
genic, J. Clin. Investig. 130 (2019), https://doi.org/10.1172/ N. Maharshak, O. Shibolet, M. Pevsner-Fischer,
JCI124196. H. Shapiro, I. Sharon, Z. Halpern, E. Segal, E. Elinav,
[241] Y. Litvak, M.X. Byndloss, A.J. Baumler, Colonocyte Post-antibiotic gut mucosal microbiome reconstitution is
metabolism shapes the gut microbiota, Science 362 impaired by probiotics and improved by autologous FMT,
(2018), https://doi.org/10.1126/science.aat9076. Cell 174 (2018) 1406e1423, https://doi.org/10.1016/
[242] A.C. Goodwin, C.E. Destefano Shields, S. Wu, D.L. Huso, j.cell.2018.08.047, e16.
X. Wu, T.R. Murray-Stewart, A. Hacker-Prietz, [247] W.R. Whitaker, E.S. Shepherd, J.L. Sonnenburg, Tunable
S. Rabizadeh, P.M. Woster, C.L. Sears, R.A.J. Casero, expression tools enable single-cell strain distinction in the
Polyamine catabolism contributes to enterotoxigenic Bac- gut microbiome, Cell 169 (2017) 538e546, https://doi.org/
teroides fragilis-induced colon tumorigenesis, Proc. Natl. 10.1016/j.cell.2017.03.041, e12.
Acad. Sci. U. S. A 108 (2011) 15354e15359, https:// [248] B. Lim, M. Zimmermann, N.A. Barry, A.L. Goodman,
doi.org/10.1073/pnas.1010203108. Engineered regulatory systems modulate gene expression
[243] K.A. Knoop, J.K. Gustafsson, K.G. McDonald, D.H. Kulkarni, of human commensals in the gut, Cell 169 (2017)
P.E. Coughlin, S. McCrate, D. Kim, C.-S. Hsieh, S.P. Hogan, 547e558, https://doi.org/10.1016/J.CELL.2017.03.045,
C.O. Elson, P.I. Tarr, R.D. Newberry, Microbial antigen e15.
encounter during a preweaning interval is critical for tolerance [249] S. Zhao, T.D. Lieberman, M. Poyet, K.M. Kauffman,
to gut bacteria, Sci. Immunol. 2 (2017), https://doi.org/ S.M. Gibbons, M. Groussin, R.J. Xavier, E.J. Alm, Adaptive
10.1126/sciimmunol.aao1314. evolution within gut microbiomes of healthy people, Cell
[244] C.E. DeStefano Shields, S.W. Van Meerbeke, Host Microbe 25 (2019) 656e667s, https://doi.org/10.1016/
F. Housseau, H. Wang, D.L. Huso, R.A.J. Casero, J.CHOM.2019.03.007, e8.

You might also like