ArticlePDF Available

Microbial Activities and Intestinal Homeostasis: A Delicate Balance Between Health and Disease

Authors:

Abstract and Figures

The concept that the intestinal microbiota modulates numerous physiological processes including immune development and function, nutrition and metabolism as well as pathogen exclusion is relatively well established in the scientific community. The molecular mechanisms driving these various effects and the events leading to the establishment of a "healthy" microbiome are slowly emerging. The objective of this review is to bring into focus important aspects of microbial/host interactions in the intestine and to discuss key molecular mechanisms controlling health and disease states. We will discuss recent evidence on how microbes interact with the host and one another and their impact on intestinal homeostasis.
Content may be subject to copyright.
REVIEW
Microbial Activities and Intestinal Homeostasis: A Delicate
Balance Between Health and Disease
Christina L. Ohland
1
and Christian Jobin
1,2
1
Department of Medicine and
2
Department of Infectious Diseases and Pathology, University of Florida, Gainesville, Florida
SUMMARY
This review discusses the equilibrium between host and
microbial community in the context of health and disease.
The focus is on bi-directional pressures between pro-
karyotes and eukaryotic cells, as well as inter-bacterial
interactions resulting in alterations to the microbiota.
The concept that the intestinal microbiota modulates
numerous physiologic processes, including immune devel-
opment and function, nutrition and metabolism, and path-
ogen exclusion, is relatively well established in the scientic
community. The molecular mechanisms driving these
various effects and the events leading to the establishment
of a healthy microbiome are slowly emerging. This review
brings into focus important aspects of microbial/host in-
teractions in the intestine and discusses key molecular
mechanisms controlling health and disease states. We
discuss the evidence of how microbes interact with the host
and one another and their impact on intestinal homeostasis.
(Cell Mol Gastroenterol Hepatol 2015;1:2840; http://
dx.doi.org/10.1016/j.jcmgh.2014.11.004)
Keywords: Bacterial Communication; Bowel Disease; Host-
Microbe Interactions; Inammatory; Microbiome.
A
ppearances can be deceiving. Despite what you see
in the mirror, humans are more prokaryotic than
eukaryotic, as the bacteria in and on our bodies outnumber
our own cells 10 to 1.
1
Microbes are embedded in our
biological system and are deeply integrated in our daily life,
and an emerging eld of research has tackled the inter-
kingdom communication network present in the walking
mixed cultures we call people.
The gastrointestinal (GI) tract has the highest density and
variety of bacteria in the human body (approximately 100
trillion microbes made up of >1000 species) due to the ideal
growth conditions provided by this organ. In the colon, there
are up to 10
12
microbes per gram of luminal content which
accounts for 60% of fecal weight.
2,3
A healthy adults intestinal
microbiome is diverse, relatively stable over time, and domi-
nated by two phyla, Bacteroidetes and Firmicutes (w95%).
Nevertheless, there is considerable interindividual variability
at the species level due to genetic, environmental, and nutri-
tional factors.
4
Diet in particular has been shown to rapidly
shift this community,
5
resulting in geographic region-specic
microbial signatures as seen in rural African children eating a
ber-rich diet compared to their European counterparts.
6
Although understanding the bacterial species present in
the gastrointestinal tract is important, recent work has shifted
the focus to the existence of a core enteric metabolome,
which has the potential to change the way we look at how our
gut functions.
7
Because bacterial genes have many over-
lapping and redundant functions, the end result of their
combined metabolism and catabolism can ultimately have a
tremendous impact on the intestinal environment and host
physiology. As we discuss later, these microbial-derived
products are a main component of the host-bacteria and
bacteria-bacteria communication network essential to intes-
tinal homeostasis.
External pressures, such as infection or antibiotics, cause
a disequilibrium in the microbial community, a phenomenon
designated as dysbiosis and often associated with pathol-
ogies including inammatory bowel disease (IBD).
8,9
Alter-
natively, internal pressure resulting from defective host
genetics, such as innate and adaptive immune genes, results
in mismanagement of the microbiota, again leading to dys-
biosis and pathologic conditions seen in the airway,
10
the
skin,
11
and the gut.
12,13
IBD is a chronic, relapsing, and
remitting inammatory disease that can be classied as
ulcerative colitis (UC) or Crohns disease (CD). Both forms
can be thought of as examples of disrupted communication
between the intestinal microbiota and the host. The nature
of this communication breakdown is not clear but involves
genetic susceptibility related to the epithelial barrier and
innate immunity, all of which are important components of
host-bacteria interaction.
14
This review will discuss the complex cross-talk between
intestinal cells and the microbiota as well as the antagonistic
and mutualistic interactions among enteric bacteria. This
multifaceted communication network is what shapes the
intestinal environment, drives homeostasis, and is thus
essential to understanding how to prevent and treat intes-
tinal disorders such as IBD.
Abbreviations used in this paper: AMP, antimicrobial peptides; CD,
Crohns disease; CDI, contact-dependent growth inhibition; GI,
gastrointestinal; HGT, horizontal gene transfer; IBD, inammatory
bowel disease; MAMP, microbe-associated molecular pattern; QS,
quorum sensing; SCFA, short-chain fatty acids; SFB, segmented la-
mentous bacteria; T6SS, type VI secretion system; UC, ulcerative
colitis.
©
2015 The Authors. Published by Elsevier Inc. on behalf of the AGA
Institute. This is an open access article under the CC BY -NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
2352-345X
http://dx.doi.org/10.1016/j.jcmgh.2014.11.004
Host Effects on the Microbiota
Gastrointestinal Tract Environment
The intestinal microbiota is largely acquired during our
rst few days of life, although recent studies have shown
that infants may not be bacteria free at birth.
15,16
Species
diversity and stability increase to an adultlike microbiota by
the age of 3 years as children are exposed to a new milieu
that includes solid food and individuals with different
microbiota.
1719
The wide topologic and geographic differ-
ences that microorganisms encounter along the GI tract
dictate their environmental niche for growth and ultimately
shape the community as a whole (Figure 1).
The acidity of the stomach limits bacterial growth and
only a sparse microbiota (<10
1
bacteria/g of contents) is
present.
20
This community predominantly consists of Fir-
micutes and Actinobacteria, but Bacteroidetes, Proteobac-
teria, and Fusobacteria are also present.
21,22
Although they
are similar at the phylum level to the rest of the GI tract, the
relative quantities of each organism differ, indicating that
the gastric population is distinct.
23
When Helicobacter pylori
are present, the stomach microbiota is dominated by this
Proteobacterium, and diversity is severely reduced.
21,23,24
In the duodenum, stomach acid is neutralized, and bile
acids become the driving force of microbiota modulation in
the small intestine. For example, cholic acid has been shown
to directly increase the cecal Firmicutes/Bacteroidetes ratio
and decrease microbial diversity in vivo.
25
Here, diversity
and bacterial load (10
3
bacteria/g) are low, but both factors
Figure 1. Regional differences in the gastrointestinal tract affect microbial niche. In the stomach, high pH and oxygen
content restrict microbial colonization. The major phyla do not signicantly differ from those found further along the GI tract,
but the predominant bacteria (Firmicutes and Actinobacteria) as well as the species are distinct. Diversity is thought to be
moderate, although this may be due to transient organisms. In the duodenum, the stomach acid is neutralized, bile is present,
oxygen is reduced by facultative anaerobes, and the host epithelium produces a mucous layer. Most starches have been
digested into monosaccharides and disaccharides, which are progressively absorbed throughout the small intestine. These
factors result in a microbiota dominated by Proteobacteria and Firmicutes such as Lactobacillales with low bacterial load and
diversity. Alternatively, host-indigestible polysaccharides promote the growth of Bacteroidetes and Firmicutes such as
Clostridiales in the colon where diversity and bacterial load are high. In addition to these factors, the intestinal barrier and
immune system affect which microbes can survive in the gut but do not appear to be region dependent.
January 2015 Microbial Activities and Homeostasis 29
increase along the length of the GI tract to the colon, which
has 10
12
bacteria/g.
20
The radial oxygen gradient with the gastrointestinal tract,
whereby oxygen diffuses inward from the epithelium into the
mucous layer, creates a microaerophilic zone in which
facultative anaerobes such as Proteobacteria thrive in an
anaerobic lumen.
26
Although nutrient absorption decreases
oxygenation of the mucosal layer by up to 40% due to
increased metabolic demands,
27
this is unlikely to substan-
tially affect the microbial community, as 4 days of hyperbaric
oxygen treatment are needed to alter the microbiota.
28
However, survival in an atmosphere with uctuating oxy-
gen does require respiratory exibility of commensal bacte-
ria. Some strict anaerobes, such as Bacteroides fragilis,
Clostridium acetobutylicum, Faecalibacterium prausnitzii,
have developed molecular switches and alternate respiratory
chains, including extracellular thiols and avins, that allow
them to tolerate low levels of oxygen over the short term and
thereby achieve a selective growth advantage.
2931
In disease states, including these accompanied by in-
testinal inammation, diminished vascular perfusion and
heightened immune cell metabolic activity result in localized
hypoxia or even anoxia within the intestines.
32
It may
therefore be surprising that intestinal dysbiosis in IBD pa-
tients is characterized by an enrichment of aerobic bacteria
including Enterobacteriaceae, which is also seen in experi-
mental models of colitis (eg, Il10
/
, Tlr5
/
, TRUC).
33,34
This disparity is puzzling but may be explained by the
tness advantage of Enterobacteriaceae, which can use ni-
trate generated during inammation as an alternative ter-
minal electron acceptor.
35
Therefore, respiration has a
fundamental inuence on the commensal bacteria that are
able to thrive in an altered gut milieu.
Dietary Components
Carbohydrates, proteins, and fats all have specic, pro-
found effects on the composition of the intestinal microbiota
and have been extensively reviewed elsewhere.
3639
Notably, Bacteroidetes and Firmicutes such as Clostridiales
are able to use host-indigestible polysaccharides and thus
dominate in the colon, whereas Proteobacteria and Firmi-
cutes such as Lactobacillales are found in the small intestine
along with high levels of monosaccharides and di-
saccharides (Figure 1).
40
Moreover, dietary vitamins can
directly affect bacterial growth, as some microbes such as
Ruminococcus bicirculans cannot synthesize their own
essential factors.
41
Dietary vitamins can also modulate the
microbiota by promoting the tness of Bacteroides species
(vitamins B
12
, C, and E) or by decreasing microbial numbers
(vitamin A, in particular for segmented lamentous
bacteria).
4244
However, it is not entirely clear whether this
is a direct effect on the bacteria or occurs indirectly via the
host epithelium or immune system, as with vitamin D.
45
In
addition to host nutrient requirements, Suez et al
46
recently
demonstrated that indigestible articial sweeteners directly
increase Bacteroidales and decrease Clostridiales in mice
and humans. This elegant study demonstrates that the
intricate relationship between our gut microbiota and what
we consume includes indigestible components and affects
whole body health.
Intestinal Immune System and Barrier Function
The gut also plays a very active role in shaping the mi-
crobial community to protect the host from the high antigenic
potential of bacterial and unwarranted immune stimulation.
The dynamic, responsive epithelial cells provide multiple
deterrents, including the formation of a mucous layer by
goblet cells, secretion of antimicrobial peptides (AMP) by
Paneth cells, intercellular tight junction complexes, and
microbe-associated molecular pattern (MAMP) recognition
systems.
4749
Similarly, the underlying immune system is
very adept at tolerating nonharmful bacteria while recog-
nizing and responding to invading pathogens and opportu-
nistic commensal organisms. Luminal sampling by
tolerogenic immune cells in the lamina propria, secretory IgA
in the mucous layer, and the complement network operate
together to maintain intestinal homeostasis.
4853
Computer-
based mathematical modeling of such a complex interaction
supports the theory that these defense systems, along with
host-derived nutrients, work together to shape the compo-
sition of the microbiota and maintain its stability.
54
Although
direct evidence of whether these systems impact microbial
species present in the intestine is lacking, host defense is a
critical factor in maintaining intestinal homeostasis.
Microbial Inuences on the Host
Educating the Immune System
Tolerance of the normal gut microbiota is an absolutely
vital element of enteric homeostasis, requiring an extensive
network of regulatory immune cells including Tregs and
tolerogenic dendritic cells.
55,56
The intestinal architecture of
the uncolonized fetal or germ-free gut is immature, with
reduced epithelial turnover, thin muscle wall and mucous
layers, a decreased number of immune cells, and disorga-
nized gut-associated lymphoid tissue.
5759
Microbial colo-
nization of mice enhances the intestinal barrier function via
MAMP signaling and educates the immune system, resulting
in maturation of the gut-associated lymphoid tissue into a
tolerant phenotype to prevent unregulated inammation
(Figure 2).
2,60
However, there appears to be a window of
opportunity for this codevelopment, as colonization of adult
germ-free mice with healthy mouse microbiota does not
induce the same protective effects as when germ-free mice
are colonized as neonates.
61,62
Segmented lamentous bacteria (SFB) are able to traverse
the mucous layer and are found attached to the small intes-
tinal epithelium of many vertebrates, including humans.
63
Although they are only present in low numbers, SFB are
essential to the development of Th17 cells through epithelial
cytokine production and dendritic cell processing,
6466
and
they are required for full immune system maturation in
mice.
67
This is an important discovery, as aberrant Th17
signaling has been documented in IBD, although evidence
that SFB have a similar function in humans is lacking.
Alternatively, Bacteroides fragilis express polysaccharide
A, which profoundly inuences the development of enteric
30 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
tolerance by converting proinammatory CD4
þ
T cells into
Tregs during colonization
68,69
and mediates equilibrium
between T
H
1 and T
H
2 responses.
70
Infants born by cesarean
delivery have decreased microbial diversity, including
delayed colonization by the B fragilis (Bacteroidetes
phylum), which is associated with decreased T
H
1 activa-
tion.
71
Similarly, strains of Clostridium clusters IV and XIVa
(Firmicutes phylum) can promote differentiation, expansion,
and colonic homing of Treg cells through bacterial antigen
signaling and short-chain fatty acids (SCFA) stimulation of
epithelial transforming growth factor-
b
1 production.
72
Thus, the far-reaching inuences of the microbiota indi-
cate the vital role of bacterial immunomodulation in main-
taining whole-body health.
Maintaining Homeostasis
Once an appropriately tolerant milieu is established,
maintenance of healthy host-microbial communication is
paramount for the host, as dysbiosis jeopardizes protective
microbial functions. A recent article by Zelante et al
73
estab-
lishes a role for microbial catabolism in balancing T-cell acti-
vation of the mucosal immune system. When converted from
sugar to tryptophan as the primary energy source, the popu-
lation of Lactobacilli expand and increase the production of
indole-3-aldehyde. This metabolite induces interleukin-22
expression through activation of aryl hydrocarbon receptors,
conferring both tolerance of the healthy microbiota and resis-
tance to the opportunistic fungal pathogen Candida albicans.
73
Figure 2. Microbial effects on the host. The microbiota induces host immune tolerance to commensal bacteria directly via a
microbe-associated molecular pattern (MAMP) and polysaccharide (PSA) signaling, indirectly through the production of short-
chain fatty acids (SCFA) and potentially through expression of epithelial intestinal alkaline phosphatase (IAP), which detoxies
luminal lipopolysaccharides (LPS). Furthermore, segmented lamentous bacteria (SFB) promote immune development of Th17
cells through epithelial cytokine production and antigen presentation by dendritic cells (DC), and the community as a whole is
required for proper gut-associated lymphoid tissue (GALT) development. SCFA also induce IgA and mucus secretion into the
lumen, promote epithelial barrier integrity, and prevent pathogen colonization. The microbiota also participates in the formation
of the active, secondary forms of bile acids.
January 2015 Microbial Activities and Homeostasis 31
Beyond regulation of the host immune system, the
microbiota inuences many other normal functions of a
healthy intestinal tract (Figure 2). For example, the micro-
biota convert bile acids into secondary forms in the lumen by
dehydroxylation, dehydrogenation, and deconjugation.
74,75
Studies in germ-free mice demonstrate that bacterial
signaling via G protein-coupled and nuclear receptors
(G protein-coupled bile acid receptor [TGR5] and farnesoid X
receptor [FXR], respectively) not only controls levels of sec-
ondary bile acids but also can modulate synthesis in the
liver.
76
Moreover, expression of intestinal alkaline phospha-
tase, an epithelial-bound enzyme that detoxies luminal
lipopolysaccharide to alleviate inammation and promote
tolerance,
77
is affected by diet and antibiotics. It is tempting to
speculate that such modulation could occur via changes in the
microbiota.
77,78
Maintaining symbiosis with our bacteria is
therefore key to preserving intestinal health and homeostasis.
The Extensive Effects of Fatty Acids
Fermentation, one of the key metabolic pathways
employed by the enteric microbiota, produces SCFA,
including acetate, propionate, and butyrate.
79
SCFA have
been shown to promote homeostatic mechanisms and pro-
tect against inammation in multiple models.
80
This topic
has been thoroughly reviewed elsewhere.
60,75,8183
Briey,
SCFA stimulate protective mucus and IgA production, pro-
mote tolerance via Treg induction, inhibit the inammatory
mediator nuclear factor kB, enhance epithelial barrier
integrity and repair, and promote competitive exclusion of
pathogens (Figure 2). However, butyrate has also been
shown to be both protective and deleterious in different
models of colorectal cancer, highlighting the complexity of
SCFA biology with a disease-specic effect.
80,84
Thus, even
predominantly benecial molecules or microbes can
potentially have negative effects in complex environments
such as the GI tract.
Microbe-Microbe Interactions
In addition to their host-mediated effect on microbial
assembly, composition, and activities, microbial niches are
also under intense pressure from other surrounding bacte-
ria. Bacteria use sophisticated intercommunication systems
to help maintain their niches; consequently, this microbial
network is essential to host homeostasis. These microbial
relationships can be antagonistic or mutualistic, depending
on the nature of the species (Figure 3). Commensal bacteria
combat other microbes using AMP production and targeted
attacks by means of specialized secretion systems, and they
Figure 3. Bacteria use a complex communication network to thrive in an environment. Bacteria interact through
combative (AC) and cooperative (DF) methods. (A) Antimicrobial peptides (AMP) typically released after cell lysis either kill or
inhibit growth of surrounding microbes. (B) Contact-dependent growth inhibition (CDI) systems deliver the toxic C-terminal end
(CdiA) into the cytoplasm of target cells after contact. (C) The type 6 secretion system (T6SS) forcefully injects toxins into
attacking bacteria. Bacteria cooperate by increasing survival of similar microbes in their vicinity with (D) horizontal gene
transfer (HGT) and (E) the formation of a protective biolm. (F) Quorum sensing (QS) allows bacteria to talk and coordinate
group behavior, and has been implicated in T6SS expression, production of AMP, and biolm formation. However, several of
these mechanisms have not been observed in the healthy human intestinal tract. Red squares represent AMP; blue triangles
are T6SS toxin; and yellow stars are QS signaling molecules.
32 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
compete for nutrients. Owing to their ruthless requirements
for survival, bacteria have also developed cooperative
mechanisms such as horizontal gene transfer, biolm for-
mation, and quorum sensing to ensure the tness of their
own community as a whole.
Combatting
Bacteria express highly potent bacteriocins, microcins,
and colicins that fend off other species or pathogens
invading their niche without causing collateral damage to
eukaryotic cells.
85
Bacteriocins are pore-forming AMP pro-
duced by Gram-positive bacteria; they directly relate to
the in vivo tness and competitiveness of Lactobacillus
salivarius and Streptococcus pneumoniae in the gut and
nasopharynx, respectively.
86,87
Microcins (<10 kDa) and
colicins (3080 kDa) are produced by Gram-negative bac-
teria, and they display a wider variety of antimicrobial
tactics than bacteriocins, including cell wall pore formation,
inhibition of RNA polymerase or tRNA synthetase, nuclease
activity, and interference with cell wall synthesis.
88,89
The degree to which the microbiota is affected by these
microbial products depends on the AMP employed. Some
have a minimal or subtle inuence in various animal models,
but others have a profound impact, such as the broad-
spectrum lacticin 3147, which drastically reduces Bacter-
oidetes and Firmicutes and increases Proteobacteria.
9092
As a similar microbial shift is also seen in IBD,
93,94
one
might speculate that bacteriocin overproduction could fos-
ter an IBD-like dysbiosis. Interestingly, mice colonized with
the bacteriocin-producing probiotic L salivarius UCC118
Bac(þ) displayed a relative increase in Bacteroidetes and
Proteobacteria compared with control mice, although no
sign of intestine inammation was reported in these mice.
95
The importance of bacterial resistance to AMP for survival
in the gut is shown in certain pathogens such as Francisella
tularensis and H pylori. These pathogens rely on the phos-
phatase LpxF to catalyze the removal of the negatively
charged 4
0
-phosphate group from the lipid A backbone, a
feature that confers a high resistance level to various
AMP.
96,97
Whether a similar system is present in commensal
bacteria is currently unknown.
Bacterial-derived AMPs integrate environmental cues
and bacterial communication in order to modulate the
microbiota composition. Expression of colicin, for example,
modulates dynamics within a simple community of three
Escherichia coli species in vitro and depends on nutrient
availability, as the carbon-storage regulator A inhibits lysis-
mediated colicin release.
98
Moreover, the colicin produced
by the pathogen Salmonella Typhimurium is up-regulated
under iron-limiting and inammatory conditions, allowing
this bacterium to outcompete other members of the Enter-
obacteriaceae family during infection.
99
However, AMP production is costly energywise, and it
often requires cell lysis for release. Therefore, bacteria have
evolved other methods for toxicity, such as contact-
dependent growth inhibition (CDI) systems or type VI
secretion systems (T6SS).
88
CDI systems are expressed by a
wide variety of Gram-negative and Gram-positive bacteria,
both pathogens and commensals.
100102
Upon contact with
another bacterium, the toxic C-terminal end of the cell
surface CdiA protein is cleaved and translocated into the
target cell where most of these family members exert
nuclease activity.
103
Expression of cognate, but not heter-
ologous, CDI proteins can prevent growth inhibition in
related species by binding the toxin, thus leading to kin
selection.
100
This type of poison-antidote tactic has been
shown to be both cooperative and combative in mathe-
matical modeling, and it likely allows for localized regula-
tion of the dynamic microbiota.
104
Interestingly, the CDI
system of Burkholderia thailandensis E264 is required for
formation of cooperative biolm communities in vitro but
acts independently of its antibacterial activity.
105,106
Conversely, T6SS systems are exclusively found in Gram-
negative bacteria and work as contractile nanomachines for
injecting toxins into target cells.
107
Remarkably, one group
demonstrated that Pseudomonas aeruginosa respond defen-
sively after being ambushed by another T6SS or a type IV
secretion system in what the investigators termed a tit-for-tat
assault, which allows them to distinguish between aggressive
and bystander T6SS-negative bacteria in vitro.
108,109
As T6SS
only target other Gram-negative bacteria or eukaryotic cells,
it follows that these injectors contribute to pathogenesis and
interbacterial competition.
110113
However, only recently
have T6SS been shown to aid in colonization of mixed bac-
terial communities by conferring a competitive advantage to
Vibrio cholera (an intestinal pathogen) and Agrobacterium
tumefaciens (Proteobacteria that infect plants).
114,115
Similar
to CDI systems, T6SS have also been implicated in the for-
mation and maintenance of biolms,
116,117
demonstrating
their versatility and potential inuence on the intestinal
microbiota.
Alternatively, pathogens can use T6SS to cause virulence.
For example, Salmonella Typhimurium requires T6SS
expression for full intracellular replication and pathogenesis
in mice.
118,119
Similarly, a clinical isolate of the diarrheagenic
Aeromonas hydrophila requires functional T6SS for viru-
lence.
120
In contrast, the T6SS of enteroaggregative E coli,an
infectious diarrheal agent associated with IBD, does not
appear to contribute to its virulence in a mouse model.
116
Competing
Carbohydrate utilization is an essential colonization
factor and modulates the dynamics of the microbiota.
121
This is the fundamental concept behind nutrient niches in
the gut and is best demonstrated by the immediate (albeit
often transient) dysbiosis that inevitably follows antibiotic
therapy.
9
Antibiotics eliminate a group of bacteria and thus
their nutrient source becomes available. This can allow new
groups of potentially pathogenic bacteria to thrive on the
previously unavailable nutrient source and, potentially,
expand to dangerous levels. This was elegantly shown by Ng
et al
122
as the postantibiotic expansion of both Salmonella
Typhimurium and Clostridium difcile in vivo depended on
their ability to use sialic acid. This monosaccharide is
January 2015 Microbial Activities and Homeostasis 33
catabolized by the microbiota but is not normally found in
the feces, as it is quickly consumed by other bacteria. After
antibiotic treatment, there is a large spike in sialic acid
availability, which returns to baseline 3 days after treatment
as the microbiota is reestablished. These pathogens exploit
the vacant nutrient niche to establish themselves in the
intestinal tract and initiate inammation.
122
Theoretically, a pathobiont could also take advantage of
increased sugar access to expand and cause pathology,
similar to how Bilophila wadsworthia blooms when supplied
with the bile acid taurocholic acid for sulte reduction.
123
Inammatory responses can further optimize the milieu
for invading species by providing an alternative method of
respiration, potentiating horizontal gene transfer of viru-
lence factors such as colicins and allowing the pathogen to
outcompete commensal bacteria.
35,99,124,125
Beyond ghting for sugars as their main carbon source,
intestinal microbes also exhibit erce competition for nitro-
gen, phosphorus, trace elements, vitamins, and other essen-
tial cofactors.
126,127
For instance, bacteria encode a wide
variety of transporters for vitamin B
12
, which indicates the
necessity of these factors for survival.
43
Moreover, the
availability of iron, zinc, and selenium have each been shown
to modulate the microbiota in vivo, theoretically by allowing
the expansion of bacteria that are more efcient at competing
for those trace elements.
128131
In fact, Salmonella Typhi-
murium infection causes up-regulation of intestinal epi-
thelialderived lipocalin-2, an AMP that interferes with
bacterial iron uptake. Sensitive bacteria are thus out-
competed by the pathogen because Salmonella Typhimurium
is resistant to lipocalin-2 action.
132
Interestingly, the pro-
biotic E coli Nissle 1917 can reduce the Salmonella Typhi-
murium burden after chronic infection has been established,
as it has multiple lipocalin-2resistant iron uptake systems
and can outcompete the pathogen for iron.
133
The ability of bacteria to metabolize host carbohydrates
and bile acids was recently shown to be critical during
mouse intestinal colonization, independent of the source of
the microbes.
134
Thus, nutrient competition is a key feature
of the microbial community dynamic and may play a role in
intestinal pathologies that exhibit nutrient deciencies. For
example, IBD is associated with deciencies in many
micronutrient levels, including zinc, vitamin A, and iron,
which could potentially inuence the underlying dysbiosis
and microbial activities.
135,136
Cooperating and Communicating
Despite the cutthroat enteric battleeld, commensal
bacteria also share byproducts rather than competing with
their neighbors for the same food source. Intricate resource
networks have evolved within the microbiota employing
this waste not, want not attitude, and they form the most
basic method of bacterial communication via detection of
neighboring microbes substrates. These networks are
beyond the scope of this article, but they have been exten-
sively reviewed elsewhere.
40,137
Another method by which bacterial cooperation occurs is
horizontal gene transfer (HGT). Specically, HGT of the
secretome is overrepresented in the intestinal microbiota, as
these molecules promote cooperation and social behavior
through the production of public goods.
138,139
One well-
established example where HGT promotes cooperation is in
the production of siderophores; iron chelators secreted to
scavenge this essential element.
140
Iron bound by side-
rophores can be taken up by any bacterium in the vicinity,
whether or not it expended the energy to create the chelator.
Thus, cheaters arise in these communities, who do not express
public goods but benet from those who do, which hinders
cooperation.
141
In limiting conditions, HGT increases the
relatedness of cheaters and thus enhances microbiota stability
by preventing the tness advantage of cheating.
142,143
Secretion of an extracellular matrix, or biolm, binds
bacteria together and protects the bacteria from outside
attacks, such as antibiotics or host immunity, while also
smothering competing microbes.
144146
Biolms aid in the
tness of a bacterial population as a whole by decreasing
the diffusion of public goods through the viscous medium
and thus preventing others from acquiring them.
147
This
function is important as biolms are especially vulnerable to
cheaters, where any reduction in biolm activity results in a
thinning of the biolm matrix and increased susceptibility to
antimicrobials.
148
These structures are not regularly found
in the bowels of healthy humans, but are thick, dense, and
adherent in untreated IBD patients.
149
This could be related
to the dysbiosis that occurs with IBD and may also explain
why many patients are refractory to antibiotic treatment
despite the apparent microbial involvement in disease.
Tying many of these cooperative pathways together is
quorum sensing (QS), the primary method by which bacteria
communicate cell to cell and coordinate group behavior.
Bacterial signaling molecules are released into the envi-
ronment and initiate gene regulation once they reach a
threshold concentration (typically in the picomolar range),
thus providing a means of functionally measuring bacteria
density.
150
QS inuences many survival mechanisms
important to intestinal bacteria, including T6SS expres-
sion,
151
antibiotic production,
126
and biolm forma-
tion.
150,152
Cheaters exist within the QS world, and these
bacteria benet from the protective signaling from other
members of the same species without expending the energy
to produce QS.
153
Similarly, some Gram-negative bacteria
eavesdrop by only expressing the QS response transcription
factor and not the signaling molecule, which could allow
commensal bacteria such as E coli to sense the presence of
invading pathogens.
154
Alternatively, some bacteria,
including multiple Proteobacteria species such as Klebsiella
pneumoniae and P aeruginosa, and even mammalian cells,
quench QS by degrading the signaling molecules to prevent
competitive colonization.
155,156
As most studies have examined pathogens, it is not
entirely clear whether human commensal bacteria use QS to
communicate despite intense speculation.
157
However, QS
signaling has been identied in the probiotics E coli Nissle
1917 and Lactobacillus species and may be involved in their
benecial effects.
158160
It is likely that bacterial communi-
cation is an important component of microbial community
assembly, composition, and activities. Further work is
34 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
needed to establish the extent to which the various forms of
microbial communication are implicated in homeostasis and
diseases.
Conclusions and Perspective
The human gut is a dynamic environment where
eukaryotic and a multitude of prokaryotic cells establish a
complex network of communication that ultimately benets
both parties. It has become clear that disrupting this pro-
ductive network has dire consequences for the host and
may contribute to intestinal pathologies, including IBD and
colorectal cancer, as well as extraintestinal disorders such
as diabetes and cardiovascular and liver diseases.
2
Although signicant progress has been made in identi-
fying host factors implicated in the disruption of the intes-
tinal microbiota and associated pathologic consequences, a
paucity of information is available about the functional
consequences of commensal microbial communication. Ge-
netic manipulation of genes implicated in these processes
(eg, QS and biolm formation in IBD) might allow dissection
of the roles of bacterial communication during health and
disease. Further advances will require the eld of microbe-
microbe communication to move from the test tube and into
the animal, as many of the fascinating interactions in vitro
have not been examined in vivo. Thus, a large piece of the
health puzzle remains missing.
Although the overwhelming majority of genes (99.1%)
examined by metagenomic sequencing of the intestinal tract
are bacterial in origin,
1
the viruses, fungi, and archaea
present may inuence specic host response and intestinal
homeostasis.
161,162
For example, susceptibility to enteric
viral infection is modulated by bacteria through enhanced
viral replication.
163
Bacteriophages in particular are an
intriguing area of research as they can enhance bacterial
tness in vivo and alter its colonization.
164,165
Although there is value in dening which bacterial species
are present in the gut, the eld is moving toward regarding
the enteric microbial community as an organ, rather than as
individual parts. For example, does a global healthy micro-
biota exist, and is it associated with a dened metabolomic
function? Because microbial metabolism is intrinsically
linked to the host, a holistic approach will be necessary to
dissect this complex relationship. Such an approach has been
successful in dening the relationship between diet, mi-
crobes, and cardiovascular diseases.
166
Although tremen-
dous progress has been made in the eld of microbiome
research, especially regarding intestinal microbiome and
IBD, key elements of the dialogues are still missing, and this
hinders the generation of novel therapeutic modalities.
Replacing an altered microbiota has been validated as a
therapeutic alternative to recurrent cases of C difcile infec-
tion,
167
but it is less clear whether IBD would be a logical
candidate for this treatment.
168
In addition, because of the
role of host genetics in IBD, it is not clear how stable and
functional a transplanted microbe would be. Consequently, a
targeted manipulation of microbial activities may represent a
better approach to disease treatment and management.
Clearly, more research is needed to identify the microbial
activities implicated in health and disease and to harness
their full potential for therapeutic purposes.
References
1. Qin J, Li R, Raes J, et al. A human gut microbial gene
catalogue established by metagenomic sequencing.
Nature 2010;464:5965.
2. Sommer F, Bäckhed F. The gut microbiotamasters of
host development and physiology. Nat Rev Microbiol
2013;11:227238.
3. Stephen AM, Cummings JH. The microbial contribution
to human faecal mass. J Med Microbiol 1980;13:45 56.
4. Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity,
stability and resilience of the human gut microbiota.
Nature 2012;489:220230.
5. David LA, Maurice CF, Carmody RN, et al. Diet rapidly
and reproducibly alters the human gut microbiome.
Nature 2014;505:559563.
6. De Filippo C, Cavalieri D, Di Paola M, et al. Impact of
diet in shaping gut microbiota revealed by a compara-
tive study in children from Europe and rural Africa. Proc
Natl Acad Sci USA 2010;107:1469114696.
7. Le Chatelier E, Nielsen T, Qin J, et al. Richness of hu-
man gut microbiome correlates with metabolic markers.
Nature 2013;500:541546.
8. Pham TAN, Lawley TD. Emerging insights on intestinal
dysbiosis during bacterial infections. Curr Opin Micro-
biol 2014;17:6774.
9. Keeney KM, Yurist-Doutsch S, Arrieta M-C, Finlay BB.
Effect of antibiotics on human microbiota and subse-
quent disease. Annu Rev Microbiol 2014;68:217235.
10. Tsicopoulos A, de Nadai P, Glineur C. Environmental
and genetic contribution in airway epithelial barrier in
asthma pathogenesis. Curr Opin Allergy Clin Immunol
2013;13:495499.
11. Zeeuwen PLJM, Kleerebezem M, Timmerman HM,
Schalkwijk J. Microbiome and skin diseases. Curr Opin
Allergy Clin Immunol 2013;13:514520.
12. Lee YJ, Park KS. Irritable bowel syndrome: emerging
paradigm in pathophysiology. World J Gastroenterol
2014;20:24562469.
13. Knights D, Lassen KG, Xavier RJ. Advances in inam-
matory bowel disease pathogenesis: linking host ge-
netics and the microbiome. Gut 2013;62:15051510.
14. Jostins L, Ripke S, Weersma RK, et al. Host-microbe
interactions have shaped the genet ic architecture of
inammatory bowel disease. N ature 2012;491:
119124.
15.
Dominguez-Bello MG, Costello EK, Contreras M, et al.
D
el
ivery mode shapes the acquisition and structure of the
initial microbiota across multiple body habitats in new-
borns. Proc Natl Acad Sci USA 2010;107:1197111975.
16. Satokari R, Grönroos T, Laitinen K, et al. Bidobacte-
rium and Lactobacillus DNA in the human placenta. Lett
Appl Microbiol 2009;48:812.
17. Bergström A, Skov TH, Bahl MI, et al. Establishment of
intestinal microbiota during early life: a longitudinal,
explorative study of a large cohort of Danish infants.
Appl Environ Microbiol 2014;80:28892900.
January 2015 Microbial Activities and Homeostasis 35
18. Koenig JE, Spor A, Scalfone N, et al. Succession of mi-
crobial consortia in the developing infant gut microbiome.
Proc Natl Acad Sci USA 2011;108(Suppl):45784585.
19. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut
microbiome viewed across age and geography. Nature
2012;486:222227.
20. Sekirov I, Russell SL, Antunes LCM, Finlay BB. Gut
microbiota in health and disease. Physiol Rev 2010;
90:859904.
21. Andersson AF, Lindberg M, Jakobsson H, et al.
Comparative analysis of human gut microbiota by bar-
coded pyrosequencing. PLoS One 2008;3:e2836.
22. Von Rosenvinge EC, Song Y, White JR, et al. Immune
status, antibiotic medication and pH are associated
with changes in the stomach uid microbiota. ISME J
2013;7:13541366.
23. Bik EM, Eckburg PB, Gill SR, et al. Molecular analysis of
the bacterial microbiota in the human stomach. Proc
Natl Acad Sci USA 2006;103:732737.
24. Cho I, Blaser MJ. The human microbiome: at the interface
of health and disease. Nat Rev Genet 2012;13:260270.
25. Islam KBMS, Fukiya S, Hagio M, et al. Bile acid is a host
factor that regulates the composition of the cecal micro-
biota in rats. Gastroenterology 2011;141:17731781.
26. Espey MG. Role of oxygen gradients in shaping redox
relationships between the human intestine and its
microbiota. Free Radic Biol Med 2013;55:130140.
27. Bohlen HG. Intestinal tissue PO2 and microvascular
responses during glucose exposure. Am J Physiol
1980;238:H164H171.
28. Albenberg L, Esipova TV, Judge CP, et al. Correlation
between intraluminal oxygen gradient and radial parti-
tioning of intestinal microbiota. Gastroenterology 2014;
147:10551063.e8.
29. Baughn AD, Malamy MH. The strict anaerobe Bacter-
oides fragilis grows in and benets from nanomolar
concentrations of oxygen. Nature 2004;427:441444.
30. Hillmann F, Fischer R-J, Saint-Prix F, et al. PerR acts as
a switch for oxygen tolerance in the strict anaerobe
Clostridium acetobutylicum. Mol Microbiol 2008;68:
848860.
31. Khan MT, Duncan SH, Stams AJM, et al. The gut
anaerobe Faecalibacterium prausnitzii uses an extra-
cellular electron shuttle to grow at oxic-anoxic in-
terphases. ISME J 2012;6:15781585
.
32. Nizet
V,
Johnson RS. Interdependence of hypoxic and
innate immune responses. Nat Rev Immunol 2009;
9:609617.
33. Kostic AD, Xavier RJ, Gevers D. The microbiome in in-
ammatory bowel disease: current status and the future
ahead. Gastroenterology 2014;146:14891499.
34. Lupp C, Robertson ML, Wickham ME, et al. Host-
mediated inammation disrupts the intestinal micro-
biota and promotes the overgrowth of Enter-
obacteriaceae. Cell Host Microbe 2007;2:119129.
35. Winter SE, Winter MG, Xavier MN, et al. Host-derived
nitrate boosts growth of E. coli in the inamed gut.
Science 2013;339:708711.
36. Albenberg LG, Wu GD. Diet and the intestinal micro-
biome: associations, functions, and implications for
health and disease. Gastroenterology 2014;
146:15641572.
37. Hamaker BR, Tuncil YE. A perspective on the complexity
of dietary ber structures and their potential effect on the
gut microbiota. J Mol Biol 2014;426:38383850.
38. Morelli L. Postnatal development of intestinal microora
as inuenced by infant nutrition. J Nutr 2008;138:
1791S1795S.
39. Walsh CJ, Guinane CM, OToole PW, Cotter PD.
Benecial modulation of the gut microbiota. FEBS Lett
2014;588:41204130.
40. Koropatkin NM, Cameron EA, Martens EC. How glycan
metabolism shapes the human gut microbiota. Nat Rev
Microbiol 2012;10:323335.
41. Wegmann U, Louis P, Goesmann A, et al. Complete
genome of a new Firmicutes species belonging to the
dominant human colonic microbiota (Ruminococcus
bicirculans) reveals two chromosomes and a selective
capacity to utilize plant glucans. Environ Microbiol
2014;16:28792890.
42. Cha H-R, Chang S-Y, Chang J-H, et al. Downregulation
of Th17 cells in the small intestine by disruption of gut
ora in the absence of retinoic acid. J Immunol 2010;
184:6799
6806.
43. Degnan
PH,
Barry NA, Mok KC, et al. Human gut mi-
crobes use multiple transporters to distinguish vitamin
B12 analogs and compete in the gut. Cell Host Microbe
2014;15:4757.
44. Siddharth J, Holway N, Parkinson SJ. A Western diet
ecological module identied from the humanized
mouse microbiota predicts diet in adults and formula
feeding in children. PLoS One 2013;8:e83689.
45. Cantorna MT, McDaniel K, Bora S, et al. Vitamin D, im-
mune regulation, the microbiota, and inammatory bowel
disease. Exp Biol Med (Maywood) 2014;239:15241530.
46. Suez J, Korem T, Zeevi D, et al. Articial sweeteners
induce glucose intolerance by altering the gut micro-
biota. Nature 2014;514:181186.
47. Hasegawa M, Inohara N. Regulation of the gut micro-
biota by the mucosal immune system in mice. Int
Immunol 2014;26:481487.
48. Hooper LV, Macpherson AJ. Immune adaptations that
maintain homeostasis with the intestinal microbiota. Nat
Rev Immunol 2010;10:159169.
49. Spasova DS, Surh CD. Blowing on embers: commensal
microbiota and our immune system. Front Immunol
2014;5:318.
50. Caricilli AM, Castoldi A, Câmara NOS. Intestinal barrier: a
gentlemens agreement between microbiota and immu-
nity. World J Gastrointest Pathophysiol 2014;5:1832.
51. Kaetzel CS. Cooperativity among secretory IgA, the
polymeric immunoglobulin receptor, and the gut
microbiota promotes hostmicrobial mutualism. Immu-
nol Lett 2014;162:1021.
52. Macpherson AJ, Geuking MB, Slack E, et al. The
habitat, double life, citizenship, and forgetfulness of IgA.
Immunol Rev 2012;245:132 146 .
53. Ricklin D, Hajishengallis G, Yang K, Lambris JD. Com-
plement: a key system for immune surveillance and
homeostasis. Nat Immunol 2010;11:785797.
36 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
54. Schluter J, Foster KR. The evolution of mutualism in gut
microbiota via host epithelial selection. PLoS Biol 2012;
10:e1001424.
55. Mann ER, Landy JD, Bernardo D, et al. Intestinal den-
dritic cells: their role in intestinal inammation, manip-
ulation by the gut microbiota and differences between
mice and men. Immunol Lett 2013;150:3040.
56. Veenbergen S, Samsom JN. Maintenance of small in-
testinal and colonic tolerance by IL-10-producing reg-
ulatory T cell subsets. Curr Opin Immunol 2012;24:
269276.
57. Bauer E, Williams BA, Smidt H, et al. Inuence of the
gastrointestinal microbiota on development of the im-
mune system in young animals. Curr Issues Intest
Microbiol 2006;7:3551.
58. Di Mauro A, Neu J, Riezzo G, et al. Gastrointestinal
function development and microbiota. Ital J Pediatr
2013;39:15.
59. Walker WA. Initial intestinal colonization in the human
infant and immune homeostasis. Ann Nutr Metab 2013;
63(Suppl 2):815.
60. Belkaid Y, Hand TW. Role of the microbiota in immunity
and inammation. Cell 2014;157:121141.
61. El Aidy S, Hooiveld G, Tremaroli V, et al. The gut
microbiota and mucosal homeostasis: colonized at
birth or at adulthood, does it matter? Gut Microbes
2013;4:118124.
62. Olszak T, An D, Zeissig S, et al. Microbial exposure
during early life has persistent effects on natural killer T
cell function. Science 2012;336:489493.
63. Schnupf P, Gaboriau-Routhiau V, Cerf-Bensussan N.
Host interactions with segmented lamentous bacteria:
an unusual trade-off that drives the post-natal matura-
tion of the gut immune system. Semin Immunol 2013;
25:342351.
64. Goto Y, Panea C, Nakato G, et al. Segmented la-
mentous bacteria antigens presented by intestinal
dendritic cells drive mucosal Th17 cell differentiation.
Immunity 2014;40:594607.
65. Ivanov II, Atarashi K, Manel N, et al. Induction of in-
testinal Th17 cells by segmented lamentous bacteria.
Cell 2009;139:485498.
66. Gaboriau-Routhiau V, Rakotobe S, Lécuyer E, et al. The
key role of segmented lamentous bacteria in the co-
ordinated maturation of gut helper T cell responses.
Immunity 2009;31:677689.
67.
Chung H, Pamp SJ, Hill JA, et al. Gut immune matu-
ration
depends
on colonization with a host-specic
microbiota. Cell 2012;149:15781593.
68. Mazmanian SK, Round JL, Kasper DL. A microbial
symbiosis factor prevents intestinal inammatory dis-
ease. Nature 2008;453:620625.
69. Round JL, Mazmanian SK. Inducible Foxp3þ regulatory
T-cell development by a commensal bacterium of the
intestinal microbiota. Proc Natl Acad Sci USA 2010;
107:1220412209.
70. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An
immunomodulatory molecule of symbiotic bacteria di-
rects maturation of the host immune system. Cell 2005;
122:107118.
71. Jakobsson HE, Abrahamsson TR, Jenmalm MC, et al.
Decreased gut microbiota diversity, delayed Bacter-
oidetes colonisation and reduced Th1 responses in in-
fants delivered by caesarean section. Gut 2014;63:
559566.
72. Atarashi K, Tanoue T, Oshima K, et al. Treg induction by
a rationally selected mixture of Clostridia strains from
the human microbiota. Nature 2013;500:232236.
73. Zelante T, Iannitti RG, Cunha C, et al. Tryptophan ca-
tabolites from microbiota engage aryl hydrocarbon re-
ceptor and balance mucosal reactivity via interleukin-
22. Immunity 2013;39:372385.
74. Jones ML, Ganopolsky JG, Martoni CJ, et al. Emerging
science of the human microbiome. Gut Microbes 2014;
5:446457.
75. Shapiro H, Thaiss CA, Levy M, Elinav E. The cross talk
between microbiota and the immune system: metabo-
lites take center stage. Curr Opin Immunol 2014;
30C:5462.
76. Sayin SI, Wahlström A, Felin J, et al. Gut microbiota
regulates bile acid metabolism by reducing the levels of
tauro-beta-muricholic acid, a naturally occurring FXR
antagonist. Cell Metab 2013;17:225235.
77. Lallès J-P. Intestinal alkaline phosphatase: novel func-
tions and protective effects. Nutr Rev 2014;72:8294.
78. Arnal M-E, Zhang J, Messori S, et al. Early changes in
microbial colonization selectively modulate intestinal
enzymes, but not inducible heat shock proteins in
young adult swine. PLoS One 2014;9:e87967.
79. Krajmalnik-Brown R, Ilhan Z-E, Kang D-W, DiBaise JK.
Effects of gut microbes on nutrient absorption and en-
ergy regulation. Nutr Clin Pract 2012;27:201214.
80. Singh N, Gurav A, Sivaprakasam S, et al. Activation of
Gpr109a, receptor for niacin and the commensal
metabolite butyrate, suppresses colonic in
ammation
and
carcinogenesis.
Immunity 2014;40:128139.
81. El Aidy S, van den Abbeele P, van de Wiele T, et al.
Intestinal colonization: how key microbial players
become established in this dynamic process: microbial
metabolic activities and the interplay between the host
and microbes. Bioessays 2013;35:913923.
82. Russell WR, Hoyles L, Flint HJ, Dumas M-E. Colonic
bacterial metabolites and human health. Curr Opin
Microbiol 2013;16:246254.
83. Thorburn AN, Macia L, Mackay CR. Diet, metabolites,
and Western-lifestyle inammatory diseases. Immu-
nity 2014;40:833842.
84. Belcheva A, Irrazabal T, Robertson SJ, et al. Gut mi-
crobial metabolism drives transformation of Msh2-
decient colon epithelial cells. Cell 2014;158:288299.
85. Ohland CL, Macnaughton WK. Probiotic bacteria and
intestinal epithelial barrier function. Am J Physiol Gas-
trointest Liver Physiol 2010;298:G807G819.
86. Dawid S, Roche AM, Weiser JN. The blp bacteriocins of
Streptococcus pneumoniae mediate intraspecies
competition both in vitro and in vivo. Infect Immun
2007;75:443451.
87. Walsh MC, Gardiner GE, Hart OM, et al. Predominance
of a bacteriocin-producing Lactobacillus salivarius
component of a ve-strain probiotic in the porcine ileum
January 2015 Microbial Activities and Homeostasis 37
and effects on host immune phenotype. FEMS Micro-
biol Ecol 2008;64:317327.
88. Braun V, Patzer SI. Intercellular communication by related
bacterial protein toxins: colicins, contact-dependent in-
hibitors, and proteins exported by the type VI secretion
system. FEMS Microbiol Lett 2013;345:1321.
89. Rebuffat S. Microcins in action: amazing defence stra-
tegies of Enterobacteria. Biochem Soc Trans 2012;
40:14561462.
90. Jozeak D, Sip A, Rawski M, et al. Dietary divercin
modies gastrointestinal microbiota and improves
growth performance in broiler chickens. Br Poult Sci
2011;52:492499.
91. Rea MC, Dobson A, OSullivan O, et al. Effect of broad-
and narrow-spectrum antimicrobials on Clostridium
difcile and microbial diversity in a model of the distal
colon. Proc Natl Acad Sci USA 2011;108(Suppl):
46394644.
92. Riboulet-Bisson E, Sturme MHJ, Jeffery IB, et al. Effect
of Lactobacillus salivarius bacteriocin Abp118 on the
mouse and pig intestinal microbiota. PLoS One 2012;
7:e31113.
93. Frank DN, St Amand AL, Feldman RA, et al. Molecular-
phylogenetic characterization of microbial community
imbalances in human inammatory bowel diseases.
Proc Natl Acad Sci USA 2007;104:1378013785.
94. Lepage P, Häsler R, Spehlmann ME, et al. Twin study
indicates loss of interaction between microbiota and
mucosa of patients with ulcerative colitis. Gastroenter-
ology 2011;141:227236.
95. Murphy EF, Cotter PD, Hogan A, et al. Divergent
metabolic outcomes arising from targeted manipulation
of the gut microbiota in diet-induced obesity. Gut 2013;
62:220226.
96. Cullen TW, Giles DK, Wolf LN, et al. Helicobacter pylori
versus the host: remodeling of the bacterial outer
membrane is required for survival in the gastric mucosa.
PLoS Pathog 2011;7:e1002454.
97. Wang X, Ribeiro AA, Guan Z, et al. Attenuated virulence
of a Francisella mutant lacking the lipid A 4
0
-phospha-
tase. Proc Natl Acad Sci USA 2007;104:41364141.
98. Hol F, Voges MJ, Dekker C, Keymer JE. Nutrient-
responsive regulation determines biodiversity in a colicin-
mediated bacterial community. BMC Biol 2014;12:68.
99. Nedialkova LP, Denzler R, Koeppel MB, et al. Inamma-
tion fuels colicin Ib-dependent competition of Salmonella
serovar Typhimurium and E. coli in enterobacterial
blooms. PLoS Pathog 2014;10:e1003844.
100. Aoki SK, DinerEJ, de Roodenbeke CT, et al. A widespread
family of polymorphic contact-dependent toxin delivery
systems in bacteria. Nature 2010;468:439442.
101. Holberger LE, Garza-Sánchez F, Lamoureux J, et al.
A novel family of toxin/antitoxin proteins in Bacillus
species. FEBS Lett 2012;586:132136.
102. Ruhe ZC, Low DA, Hayes CS. Bacterial contact-
dependent growth inhibition. Trends Microbiol 2013;
21:230237.
103. Webb JS, Nikolakakis KC, Willett JLE, et al. Delivery of
CdiA nuclease toxins into target cells during contact-
dependent growth inhibition. PLoS One 2013;8:e57609.
104. Blanchard AE, Celik V, Lu T. Extinction, coexistence,
and localized patterns of a bacterial population
with contact-dependent inhibition. BMC Syst Biol 2014;
8:23.
105. Anderson MS, Garcia EC, Cotter PA. Kind discrimina-
tion and competitive exclusion mediated by contact-
dependent growth inhibition systems shape biolm
community structure. PLoS Pathog 2014;10:e1004076.
106. Garcia EC, Anderson MS, Hagar JA, Cotter PA. Bur-
kholderia BcpA mediates biolm formation indepen-
dently of interbacterial contact-dependent growth
inhibition. Mol Microbiol 2013;89:12131225.
107. Kapitein N, Mogk A. Deadly syringes: type VI secretion
system activities in pathogenicity and interbacterial
competition. Curr Opin Microbiol 2013;16:5258.
108. Basler M, Ho BT, Mekalanos JJ. Tit-for-tat: type VI
secretion system counterattack during bacterial cell-cell
interactions. Cell 2013;152:884894.
109. Ho BT, Basler M, Mekalanos JJ. Type 6 secretion
system-mediated immunity to type 4 secretion system-
mediated gene transfer. Science 2013;342:250253.
110. Hood RD, Singh P, Hsu F, et al. A type VI secretion
system of Pseudomonas aeruginosa targets a toxin to
bacteria. Cell Host Microbe 2010;7:2537.
111.
Russell AB, Wexler AG, Harding BN, et al. A type VI
secretion-related
pathway
in bacteroidetes mediates
interbacterial antagonism. Cell Host Microbe 2014;16:
227236.
112. Schwarz S, West TE, Boyer F, et al. Burkholderia type VI
secretion systems have distinct roles in eukaryotic and
bacterial cell interactions. PLoS Pathog 2010;6:
e1001068.
113. Unterweger D, Miyata ST, Bachmann V, et al. The Vibrio
cholerae type VI secretion system employs diverse
effector modules for intraspecic competition. Nat
Commun 2014;5:3549.
114. Fu Y, Waldor MK, Mekalanos JJ. Tn-Seq analysis of
Vibrio cholerae intestinal colonization reveals a role for
T6SS-mediated antibacterial activity in the host. Cell
Host Microbe 2013;14:652663.
115. Ma L-S, Hachani A, Lin J-S, et al. Agrobacterium
tumefaciens deploys a superfamily of type VI secretion
DNase effectors as weapons for interbacterial compe-
tition in planta. Cell Host Microbe 2014;16:94 104 .
116. Aschtgen M-S, Bernard CS, de Bentzmann S, et al.
SciN is an outer membrane lipoprotein required for type
VI secretion in enteroaggregative Escherichia coli.
J Bacteriol 2008;190:75237531.
117. de Pace F, Boldrin de Paiva J, Nakazato G, et al.
Characterization of IcmF of the type VI secretion system
in an avian pathogenic Escherichia coli (APEC) strain.
Microbiology 2011;157:29542962.
118. Mulder DT, Cooper CA, Coombes BK. Type VI secretion
system-associated gene clusters contribute to patho-
genesis of Salmonella enterica serovar Typhimurium.
Infect Immun 2012;80:1996 2007.
119. Parsons DA, Heffron F. sciS, an icmF homolog in Sal-
monella enterica serovar Typhimurium, limits intracel-
lular replication and decreases virulence. Infect Immun
2005;73:43384345.
38 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
120. Suarez G, Sierra JC, Sha J, et al. Molecular character-
ization of a functional type VI secretion system from a
clinical isolate of Aeromonas hydrophila. Microb Pathog
2008;44:344361.
121. Lee SM, Donaldson GP, Mikulski Z, et al. Bacterial
colonization factors control specicity and stability of
the gut microbiota. Nature 2013;501:426429.
122. Ng KM, Ferreyra JA, Higginbottom SK, et al. Microbiota-
liberated host sugars facilitate post-antibiotic expansion
of enteric pathogens. Nature 2013;502:9699.
123. Devkota S, Wang Y, Musch MW, et al. Dietary-fat-
induced taurocholic acid promotes pathobiont expan-
sion and colitis in Il10
/
mice. Nature 2012;487:
104108.
124. Winter SE, Thiennimitr P, Winter MG, et al. Gut inam-
mation provides a respiratory electron acceptor for
Salmonella. Nature 2010;467:426429.
125. Stecher B, Robbiani R, Walker AW, et al. Salmonella
enterica serovar Typhimurium exploits inammation to
compete with the intestinal microbiota. PLoS Biol 2007;
5:21772189.
126. Hibbing ME, Fuqua C, Parsek MR, Peterson SB. Bac-
terial competition: surviving and thriving in the microbial
jungle. Nat Rev Microbiol 2010;8:1525.
127. Seth EC, Taga ME. Nutrient cross-feeding in the mi-
crobial world. Front Microbiol 2014;5:350.
128. Gielda LM, DiRita VJ. Zinc competition among the in-
testinal microbiota. MBio 2012;3:e00171e00212.
129. Kasaikina MV, Kravtsova MA, Lee BC, et al. Dietary se-
lenium affects host selenoproteome expression by inu-
encing the gut microbiota. FASEB J 2011;25:24922499.
130. Tompkins GR, ODell NL, Bryson IT, Pennington CB.
The effects of dietary ferric iron and iron deprivation on
the bacterial composition of the mouse intestine. Curr
Microbiol 2001;43:3842.
131. Werner T, Wagner SJ, Martínez I, et al. Depletion of
luminal iron alters the gut microbiota and prevents
Crohns disease-like ileitis. Gut 2011;60:325333.
132. Raffatellu M, George MD, Akiyama Y, et al. Lipocalin-2
resistance confers an advantage to Salmonella enterica
serotype Typhimurium for growth and survival in the
inamed intestine. Cell Host Microbe 2009;5:476486.
133. Deriu E, Liu JZ, Pezeshki M, et al. Probiotic bacteria
reduce Salmonella Typhimurium intestinal colonization
by competing for iron. Cell Host Microbe 2013;14:2637.
134. Seedorf H, Grifn NW, Ridaura VK, et al. Bacteria from
diverse habitats colonize and compete in the mouse
gut. Cell 2014;159:253266.
135. Alkhouri RH, Hashmi H, Baker RD, et al. Vitamin and
mineral status in patients with in
ammatory bowel dis-
ease.
J
Pediatr Gastroenterol Nutr 2013;56:8992.
136. Guagnozzi D, Lucendo AJ. Anemia in inammatory
bowel disease: a neglected issue with relevant effects.
World J Gastroenterol 2014;20:35423551.
137. Fischbach MA, Sonnenburg JL. Eating for two: how
metabolism establishes interspecies interactions in the
gut. Cell Host Microbe 2011;10:336347.
138. Dimitriu T, Lotton C, Benard-Capelle J, et al. Genetic
information transfer promotes cooperation in bacteria.
Proc Natl Acad Sci USA 2014;111:1110311108.
139. Nogueira T, Rankin DJ, Touchon M, et al. Horizontal gene
transfer of the secretome drives the evolution of bacterial
cooperation and virulence. Curr Biol 2009;19:16831691.
140. Grifn AS, West SA, Buckling A. Cooperation and
competition in pathogenic bacteria. Nature 2004;430:
10241027.
141. Harrison F, Buckling A. Hypermutability impedes
cooperation in pathogenic bacteria. Curr Biol 2005;
15:19681971.
142. Jousset A, Eisenhauer N, Materne E, Scheu S. Evolu-
tionary history predicts the stability of cooperation in
microbial communities. Nat Commun 2013;4:2573.
143. Mc Ginty SÉ, Lehmann L, Brown SP, Rankin DJ. The
interplay between relatedness and horizontal gene
transfer drives the evolution of plasmid-carried public
goods. Proc Biol Sci 2013;280:20130400.
144. Nadell CD, Bassler BL. A tness trade-off between local
competition and dispersal in Vibrio cholerae biolms.
Proc Natl Acad Sci USA 2011;108:1418114185.
145. Vlamakis H, Chai Y, Beauregard P, et al. Sticking
together: building a biolm the Bacillus subtilis way. Nat
Rev Microbiol 2013;11:157168.
146. Xavier JB, Foster KR. Cooperation and conict in mi-
crobial biolms. Proc Natl Acad Sci USA 2007;104:
876881.
147. Drescher K, Nadell CD, Stone HA, et al. Solutions to the
public goods dilemma in bacterial biolms. Curr Biol
2014;24:5055
.
148. Popat
R,
Crusz SA, Messina M, et al. Quorum-sensing
and cheating in bacterial biolms. Proc Biol Sci 2012;
279:47654771.
149. Swidsinski A, Weber J, Loening-Baucke V, et al. Spatial
organization and composition of the mucosal ora in
patients with inammatory bowel disease. J Clin
Microbiol 2005;43:33803389.
150. Williams P, Winzer K, Chan WC, Cámara M. Look whos
talking: communication and quorum sensing in the
bacterial world. Philos Trans R Soc Lond B Biol Sci
2007;362:11191134.
151. Zheng J, Shin OS, Cameron DE, Mekalanos JJ. Quorum
sensing and a global regulator TsrA control expression
of type VI secretion and virulence in Vibrio cholerae.
Proc Natl Acad Sci USA 2010;107:2112821133.
152. Dickschat JS. Quorum sensing and bacterial bio lms.
Nat Prod Rep 2010;27:343369 .
153. Sandoz KM, Mitzimberg SM, Schuster M. Social
cheating in Pseudomonas aeruginosa quorum sensing.
Proc Natl Acad Sci USA 2007;104:1587615881.
154. Swearingen MC, Sabag-Daigle A, Ahmer BMM. Are
there acyl-homoserine lactones within mammalian in-
testines? J Bacteriol 2013;195:173 179 .
155. Hong K-W, Koh C-L, Sam C-K, et al. Quorum
quenching revisitedfrom signal decays to signalling
confusion. Sensors (Basel) 2012;12:46614696.
156. Kalia VC. Quorum sensing inhibitors: an overview.
Biotechnol Adv 2013;31:224245.
157. Lukás F, Gorenc G, Kopecný J. Detection of possible
AI-2-mediated quorum sensing system in commensal
intestinal bacteria. Folia Microbiol (Praha) 2008;53:
221224.
January 2015 Microbial Activities and Homeostasis 39
158. Jacobi CA, Grundler S, Hsieh C-J, et al. Quorum
sensing in the probiotic bacterium Escherichia coli
Nissle 1917 (Mutaor)evidence that furanosyl borate
diester (AI-2) is inuencing the cytokine expression in
the DSS colitis mouse model. Gut Pathog 2012;4:8.
159. Lebeer S, De Keersmaecker SCJ, Verhoeven TLA, et al.
Functional analysis of luxS in the probiotic strain
Lactobacillus rhamnosus GG reveals a central meta-
bolic role important for growth and biolm formation.
J Bacteriol 2007;189:860871.
160. Van Hemert S, Meijerink M, Molenaar D, et al. Identi-
cation of Lactobacillus plantarum genes modulating the
cytokine response of human peripheral blood mono-
nuclear cells. BMC Microbiol 2010;10:293.
161. Iliev ID, Funari VA, Taylor KD, et al. Interactions between
commensal fungi and the C-type lectin receptor Dectin-
1inuence colitis. Science 2012;336:13141317.
162. Norman JM, Handley SA, Virgin HW. Kingdom-agnostic
metagenomics and the importance of complete char-
acterization of enteric microbial communities. Gastro-
enterology 2014;146:14591469.
163. Kuss SK, Best GT, Etheredge CA, et al. Intestinal
microbiota promote enteric virus replication and sys-
temic pathogenesis. Science 2011;334:249252.
164. Duerkop BA, Clements CV, Rollins D, et al. A composite
bacteriophage alters colonization by an intestinal
commensal bacterium. Proc Natl Acad Sci USA 2012;
109:1762117626.
165. Reyes A, Wu M, McNulty NP, et al. Gnotobiotic mouse
model of phage-bacterial host dynamics in the human
gut. Proc Natl Acad Sci USA 2013;110:2023620241.
166. Tang WHW, Hazen SL. The contributory role of gut
microbiota in cardiovascular disease. J Clin Invest
2014;124:42044211.
167. Lo Vecchio A, Cohen MB. Fecal microbiota trans-
plantation for Clostridium difcile infection: benets and
barriers. Curr Opin Gastroenterol 2014;30:4753.
168. Colman RJ, Rubin DT. Fecal microbiota transplantation
as therapy for inammatory bowel disease: a system-
atic review and meta-analysis. J Crohns Colitis 2014;8:
15691581
.
Received October 21, 2014. Accepted November 20, 2014.
Correspondence
Address correspondence to: Christian Jobin, PhD, Department of Medicine,
University of Florida, 2033 Mowry Road, Ofce 461, Gainesville, Florida
32610. e-mail: christian.jobin@medicine.u.edu; fax: (352) 392-3944.
Conicts of interest
The authors disclose no conicts.
Funding
This work was supported by the National Institutes of Health (RO1DK047700
and RO1DK073338 to C.J.).
40 Ohland and Jobin Cellular and Molecular Gastroenterology and Hepatology Vol. 1, No. 1
... [3] Gut microbes are gaining importance due to their functions of stimulating host immune development, nutrient metabolism, differentiation of mucosal structure, [3] maintaining intestinal mucosal barrier by enhancing gut integrity, formation of the intestinal epithelium, and protection against pathogens, providing anti-inflammatory signals to the host, [2] and are necessary for the maintenance of intestinal homeostasis. [5] The role of microbiomes is crucial during early life as the changes in relation to the composition and the abundance of microbiomes become more or less stable, remain the same throughout life, and dictate the health of the host. [5] Various internal and external factors influence the gut microbiota, starting from birth to adulthood. ...
... [5] The role of microbiomes is crucial during early life as the changes in relation to the composition and the abundance of microbiomes become more or less stable, remain the same throughout life, and dictate the health of the host. [5] Various internal and external factors influence the gut microbiota, starting from birth to adulthood. [3] The human fecal microbiota consists of four main groups of bacteria (phyla), that are Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria, of which the first two bacteria account for more than 80% of the microbiota. ...
Article
Objectives Antibiotic use is associated with dysbiosis of the gut microbiome. The objective of this study is to investigate the effect of antibiotics on gut composition in children aged 3–5 years receiving antibiotics compared to children who did not receive antibiotics. Materials and Methods A total of 54 participants aged 3–5 years were included in this multi-centric cohort study. Participants were divided into two equal groups, that is the treatment-experienced group (Group 1, n = 27, antibiotic) and the treatment-not-experienced group (Group 2, n = 27, non-antibiotic). Stool samples of study participants were collected on days 0 and 5 (± 1 day) and analyzed using 16Svedberg ribosomal ribonucleic acid (16S rRNA) gene sequencing. Statistical Analysis The Kruskal-Wallis H-test and Benjamini-Hochberg FDR correction were applied to determine the differentially abundant pathways across the zones using Statistical Analysis of Metagenomic Profiles (STAMP) (v2.1.3). Results A non-significant increase in the mean abundance of the Phyla Bacteroidota, Proteobacteria, Actinobacteria , and Verrucomicrobiota was observed in both groups from day 0 to day 5. An alteration in the Firmicutes / Bacteroidetes ratio was observed. A significant ( P < 0.05) abundance of genus Enterobacteriaceae, Enterococcaceae , and Lactobacillaceae was observed in the participants with antibiotic treatment. The relative abundance of families Enterococcus, Lactobacillus, Sellimonas, Ruminococcus, Torques , and Eggerthella groups was observed to be significantly higher ( P < 0.05) in participants with antibiotic treatment. Beta-diversity indices revealed significant differences at group and subgroup levels regarding the bacterial counts. Conclusions It was observed that a short-term course of 5 days of antibiotic usage is associated with altered microbial abundance and diversity.
... ENaC is crucial for sodium absorption in various tissues, including the kidneys and gastrointestinal tract [66,67]. An increased presence or activity of SCNN1B and SCNN1G could potentially reflect the body's effort to maintain sodium homeostasis in the face of high dietary sodium [68,69]. The lack of these ENaC genes in LSD (but not HSD) could be part of a compensatory mechanism that is less active or unnecessary when the sodium intake is lower [70]. ...
Article
Full-text available
The gut microbiota is a dynamic ecosystem that plays a pivotal role in maintaining host health. The perturbation of these microbes has been linked to several health conditions. Hence, they have emerged as promising targets for understanding and promoting good health. Despite the growing body of research on the role of sodium in health, its effects on the human gut microbiome remain under-explored. Here, using nutrition and metagenomics methods, we investigate the influence of dietary sodium intake and alterations of the human gut microbiota. We found that a high-sodium diet (HSD) altered the gut microbiota composition with a significant reduction in Bacteroides and inverse increase in Prevotella compared to a low-sodium diet (LSD). However, there is no clear distinction in the Firmicutes/Bacteroidetes (F/B) ratio between the two diet types. Metabolic pathway reconstruction revealed the presence of sodium reabsorption genes in the HSD, but not LSD. Since it is currently difficult in microbiome studies to confidently associate the F/B ratio with what is considered healthy (e.g., low sodium) or unhealthy (e.g., high sodium), we suggest that the use of a genus-based ratio such as the Bacteroides/Prevotella (B/P) ratio may be more beneficial for the application of microbiome studies in health.
... Our results in the lactobacillus-treated group were in line with the results obtained by Pooljun et al. (2020), who found that hepatopancreas of shrimp fed on lactobacillus then challenged with vibrio having normal structure and fewer pathological alterations occur. These findings were attributed to the action of the polysaccharides (such as α-D-glucan and β-D-glucan) of the yeast cell wall that binds to the bacterial outer membrane, preventing the attachment and colonization of pathogens in the gastrointestinal tract (Ohland and Jobin 2015). ...
... The intestinal tract (gut) of humans serves as an important habitat for diverse microbial commensals, also known as microbiota colonizing and adapting to the gut environment creating a state of homeostasis [1,2]. Microbiota colonization of the gut begins soon after childbirth and continues throughout one's lifetime depending on the immediate ecosystem they belong to and their overall lifestyle. ...
Article
Full-text available
Background Cholera can result in the expulsion of important microbiota from the gut and result in death if left untreated. The disease transmits mainly via drinking water carrying Vibrio cholerae; and household contacts (HHC) of cholera patients are at elevated risk during the first week of infection. The gut microbiota profiles of HHC-children of cholera patients at Dhaka city slums were investigated before (day 0) and after (day 8) delivery of chlorinated water as part of the major study ‘CHoBI7 trial (cholera-hospital-based intervention for 7 days)’. Result Results of sequencing and analysis of bacterial community DNA revealed the predominance of two bacterial phyla: Bacteroidetes and Firmicutes at day 0 with a relative abundance of 62 ± 6 (mean ± SEM%) and 32 ± 7, respectively. The pattern reversed at day 8 with a decreased relative abundance of Bacteroidetes (39 ± 12; p = 0.034) and an increased abundance of Firmicutes (49 ± 12; p = 0.057). Of 65 bacterial families confirmed at day 0, six belonging to Proteobacteria including Vibrionaceae disappeared at day 8. Interestingly, the relative abundance of four Firmicutes families—Lachnospiraceae, Bifidobacteriaceae, Clostridiaceae, and Ruminococcaceae was increased in all five study children at day 8. Conclusion The observed exclusion of pathogenic Proteobacteria and enhancement of beneficial Firmicutes in the gut of children delivered with chlorinated water as part of WASH intervention reflect a great promise of the CHoBI7 program in preventing cholera and improving child health.
... The community of microbes performs a variety of tasks for the host, including facilitating the uptake of nutrients (Martin et al., 2019), preserving homeostasis (Ohland and Jobin, 2015), fending off pathogens (Pickard et al., 2017), regulating immunological response (Mendes et al., 2019), among many others. Understanding these tasks and revealing the dialog between the bacterium and the host may help in developing plans for preserving the health status, treating diseases. ...
Article
Full-text available
Numerous biological environments have been characterized with the advent of metagenomic sequencing using next generation sequencing which lays out the relative abundance values of microbial taxa. Modeling the human microbiome using machine learning models has the potential to identify microbial biomarkers and aid in the diagnosis of a variety of diseases such as inflammatory bowel disease, diabetes, colorectal cancer, and many others. The goal of this study is to develop an effective classification model for the analysis of metagenomic datasets associated with different diseases. In this way, we aim to identify taxonomic biomarkers associated with these diseases and facilitate disease diagnosis. The microBiomeGSM tool presented in this work incorporates the pre-existing taxonomy information into a machine learning approach and challenges to solve the classification problem in metagenomics disease-associated datasets. Based on the G-S-M (Grouping-Scoring-Modeling) approach, species level information is used as features and classified by relating their taxonomic features at different levels, including genus, family, and order. Using four different disease associated metagenomics datasets, the performance of microBiomeGSM is comparatively evaluated with other feature selection methods such as Fast Correlation Based Filter (FCBF), Select K Best (SKB), Extreme Gradient Boosting (XGB), Conditional Mutual Information Maximization (CMIM), Maximum Likelihood and Minimum Redundancy (MRMR) and Information Gain (IG), also with other classifiers such as AdaBoost, Decision Tree, LogitBoost and Random Forest. microBiomeGSM achieved the highest results with an Area under the curve (AUC) value of 0.98% at the order taxonomic level for IBDMD dataset. Another significant output of microBiomeGSM is the list of taxonomic groups that are identified as important for the disease under study and the names of the species within these groups. The association between the detected species and the disease under investigation is confirmed by previous studies in the literature. The microBiomeGSM tool and other supplementary files are publicly available at: https://github.com/malikyousef/microBiomeGSM .
... T he metabolic activities of microorganisms are the basis for all life on Earth. They drive biogeochemical cycles, influence eukaryotic hosts, remediate contaminated sites, and play an important role in biotechnology (1)(2)(3)(4). However, our current knowledge of microbial in situ activity is still mainly based on bulk analyses of microbiomes as well as deductions from cultivated isolates and laboratory enrichments that are considered representative (5). ...
Article
Full-text available
Current methods for the identification of specific microorganisms based on an in situ metabolism are often hampered by insufficient sensitivity and habitat complexity. Here, we present a novel approach for identifying and sequencing single microbial cells metabolizing a specific organic compound with high sensitivity and without prior knowledge of the microbial community. The workflow consists of labeling individual cells with a [¹⁴C] substrate based on their metabolic activity, followed by encapsulating cells in alginate with nuclear emulsion by using microfluidics. We here adapted the concept of microautoradiography to visually distinguish between encapsulated labeled and non-labeled cells, which are then sorted via flow cytometry for single cell genomics. As a proof-of-concept, we labeled, separated, lysed, and sequenced single cells of the benzene degrader Pseudomonas veronii from mock microbial communities. The cells of P. veronii were isolated with 100% specificity. Single-cell microautoradiography and genome sequencing is an innovative method for elucidating microbial identity, activity, and function in diverse habitats, contributing to elucidate novel taxa and genes with potential for biotechnological applications such as bioremediation. IMPORTANCE A central question in microbial ecology is which member of a community performs a particular metabolism. Several sophisticated isotope labeling techniques are available for analyzing the metabolic function of populations and individual cells in a community. However, these methods are generally either insufficiently sensitive or throughput-limited and thus have limited applicability for the study of complex environmental samples. Here, we present a novel approach that combines highly sensitive radioisotope tracking, microfluidics, high-throughput sorting, and single-cell genomics to simultaneously detect and identify individual microbial cells based solely on their in situ metabolic activity, without prior information on community structure.
Article
The intestinal immune system plays a crucial role in obesity and insulin resistance. An altered intestinal immunity is associated with changes to the gut microbiota, barrier function, and tolerance to luminal antigens. Lipid metabolism and its unbalance can also contribute to acute and chronic inflammation in different conditions. In celiac disease (CD), the serum phospholipid profile in infants who developed CD is dramatically different when compared to that of infants at risk of CD not developing the disease. In a mouse model of gluten sensitivity, oral wheat gliadin challenge in connection with inhibition of the metabolism of arachidonic acid, an omega‐6 polyunsaturated fatty acid, specifically induces the enteropathy. Recent evidence suggests that gluten may play a role also for development of life‐style related diseases in populations on a high fat diet (HFD). However, the mechanisms behind these effects are not yet understood. Exploratory studies in mice feed HFD show that wheat gliadin consumption affects glucose and lipid metabolic homeostasis, alters the gut microbiota, and the immune cell profile in liver.
Article
The human gut microbiota regulates estrogen metabolism through the “estrobolome,” the collection of bacterial genes that encode enzymes like β‐glucuronidases and β‐glucosidases. These enzymes deconjugate and reactivate estrogen, influencing circulating levels. The estrobolome mediates the enterohepatic circulation and bioavailability of estrogen. Alterations in gut microbiota composition and estrobolome function have been associated with estrogen‐related diseases like breast cancer, enometrial cancer, and polycystic ovarian syndrome (PCOS). This is likely due to dysregulated estrogen signaling partly contributed by the microbial impacts on estrogen metabolism. Dietary phytoestrogens also undergo bacterial metabolism into active metabolites like equol, which binds estrogen receptors and exhibits higher estrogenic potency than its precursor daidzein. However, the ability to produce equol varies across populations, depending on the presence of specific gut microbes. Characterizing the estrobolome and equol‐producing genes across populations can provide microbiome‐based biomarkers. Further research is needed to investigate specific components of the estrobolome, phytoestrogen‐microbiota interactions, and mechanisms linking dysbiosis to estrogen‐related pathology. However, current evidence suggests that the gut microbiota is an integral regulator of estrogen status with clinical relevance to women's health and hormonal disorders.
Article
The early provision of soluble/insoluble fiber to the patient who is critically ill has been controversial in the past. Especially in the setting of hemodynamic instability, dysmotility, or impaired gastrointestinal transit, fear of inspissation of formula with precipitation of nonocclusive mesenteric ischemia (NOMI)/nonocclusive bowel necrosis (NOBN) limited its utilization by medical and surgical intensivists. The incidence of NOMI/NOBN has been estimated at 0.2%–0.3% for all intensive care unit (ICU) patients receiving enteral nutrition (EN), and the occurrence of inspissated formula is even less. The science supporting a benefit from providing fiber has recently increased exponentially. The fermentation of soluble fibers leading to the production of short chain fatty acids supports gut barrier function, modulates immune responses, and promotes refaunation of commensal organisms. The “butyrate effect” refers to local (gastrointestinal tract) and systemic anti‐inflammatory responses mediated by the M2 polarization of macrophages, inhibition of histone deacetylase, and stimulation of ubiquitous G protein receptors. Both soluble and insoluble fiber have been shown to promote intestinal motility, reduce feeding intolerance, and shorten hospital length of stay. The benefit of providing dietary fiber early upon admission to the ICU outweighs its minimal associated risk. The point at which the intensivist determines that is safe to initiate EN, both soluble and insoluble fiber should be included in the enteral formulation.
Article
Full-text available
Humans host complex microbial communities believed to contribute to health maintenance and, when in imbalance, to the development of diseases. Determining the microbial composition in patients and healthy controls may thus provide novel therapeutic targets. For this purpose, high-throughput, cost-effective methods for microbiota characterization are needed. We have employed 454-pyrosequencing of a hyper-variable region of the 16S rRNA gene in combination with sample-specific barcode sequences which enables parallel in-depth analysis of hundreds of samples with limited sample processing. In silico modeling demonstrated that the method correctly describes microbial communities down to phylotypes below the genus level. Here we applied the technique to analyze microbial communities in throat, stomach and fecal samples. Our results demonstrate the applicability of barcoded pyrosequencing as a high-throughput method for comparative microbial ecology.
Article
Full-text available
To study how microbes establish themselves in a mammalian gut environment, we colonized germ-free mice with microbial communities from human, zebrafish, and termite guts, human skin and tongue, soil, and estuarine microbial mats. Bacteria from these foreign environments colonized and persisted in the mouse gut; their capacity to metabolize dietary and host carbohydrates and bile acids correlated with colonization success. Cohousing mice harboring these xenomicrobiota or a mouse cecal microbiota, along with germ-free "bystanders," revealed the success of particular bacterial taxa in invading guts with established communities and empty gut habitats. Unanticipated patterns of ecological succession were observed; for example, a soil-derived bacterium dominated even in the presence of bacteria from other gut communities (zebrafish and termite), and human-derived bacteria colonized germ-free bystander mice before mouse-derived organisms. This approach can be generalized to address a variety of mechanistic questions about succession, including succession in the context of microbiota-directed therapeutics.
Article
Full-text available
Non-caloric artificial sweeteners (NAS) are among the most widely used food additives worldwide, regularly consumed by lean and obese individuals alike. NAS consumption is considered safe and beneficial owing to their low caloric content, yet supporting scientific data remain sparse and controversial. Here we demonstrate that consumption of commonly used NAS formulations drives the development of glucose intolerance through induction of compositional and functional alterations to the intestinal microbiota. These NAS-mediated deleterious metabolic effects are abrogated by antibiotic treatment, and are fully transferrable to germ-free mice upon faecal transplantation of microbiota configurations from NAS-consuming mice, or of microbiota anaerobically incubated in the presence of NAS. We identify NAS-altered microbial metabolic pathways that are linked to host susceptibility to metabolic disease, and demonstrate similar NAS-induced dysbiosis and glucose intolerance in healthy human subjects. Collectively, our results link NAS consumption, dysbiosis and metabolic abnormalities, thereby calling for a reassessment of massive NAS usage.
Article
Full-text available
Background Antagonistic interactions mediated by antibiotics are strong drivers of bacterial community dynamics which shape biodiversity. Colicin production by Escherichia coli is such an interaction that governs intraspecific competition and is involved in promoting biodiversity. It is unknown how environmental cues affect regulation of the colicin operon and thus influence antibiotic-mediated community dynamics.ResultsHere, we investigate the community dynamics of colicin-producing, -sensitive, and -resistant/non-producer E. coli strains that colonize a microfabricated spatially-structured habitat. Nutrients are found to strongly influence community dynamics: when growing on amino acids and peptides, colicin-mediated competition is intense and the three strains do not coexist unless spatially separated at large scales (millimeters). Surprisingly, when growing on sugars, colicin-mediated competition is minimal and the three strains coexist at the micrometer scale. Carbon storage regulator A (CsrA) is found to play a key role in translating the type of nutrients into the observed community dynamics by controlling colicin release. We demonstrate that by mitigating lysis, CsrA shapes the community dynamics and determines whether the three strains coexist. Indeed, a mutant producer that is unable to suppress colicin release, causes the collapse of biodiversity in media that would otherwise support co-localized growth of the three strains.Conclusions Our results show how the environmental regulation of an antagonistic trait shapes community dynamics. We demonstrate that nutrient-responsive regulation of colicin release by CsrA, determines whether colicin producer, resistant non-producer, and sensitive strains coexist at small spatial scales, or whether the sensitive strain is eradicated. This study highlights how molecular-level regulatory mechanisms that govern interference competition give rise to community-level biodiversity patterns.
Article
Full-text available
Vertebrates have co-evolved with microorganisms resulting in a symbiotic relationship, which plays an important role in health and disease. Skin and mucosal surfaces are colonized with a diverse population of commensal microbiota, over 1000 species, outnumbering the host cells by 10-fold. In the past 40 years, studies have built on the idea that commensal microbiota is in constant contact with the host immune system and thus influence immune function. Recent studies, focusing on mutualism in the gut, have shown that commensal microbiota seems to play a critical role in the development and homeostasis of the host immune system. In particular, the gut microbiota appears to direct the organization and maturation of lymphoid tissues and acts both locally and systemically to regulate the recruitment, differentiation, and function of innate and adaptive immune cells. While the pace of research in the area of the mucosal-immune interface has certainly intensified over the last 10 years, we are still in the early days of this field. Illuminating the mechanisms of how gut microbes shape host immunity will enhance our understanding of the causes of immune-mediated pathologies and improve the design of next-generation vaccines. This review discusses the recent advances in this field, focusing on the close relationship between the adaptive immune system and commensal microbiota, a constant and abundant source of foreign antigens.
Article
Full-text available
Significance Bacteria often cooperate through the production of public goods that change their environment. These processes can affect human health by increasing virulence or antibiotic resistance. Public good production is costly, making cooperation susceptible to invasion by nonproducing “cheater” individuals. Bacteria also readily share genes, even among distinct species. Our experiments and models converge to show that when both cheating and cooperative genes are transferred, cooperators win against cheaters because transfer increases assortment among alleles, favoring cooperation. This can explain why genes for cooperation are often mobile, and suggests that, in addition to reducing antibiotic resistance spread, preventing gene mobility could reduce cooperative virulence.
Article
The US National Institute of Health-funded Human Microbiome Project Consortium started 6 years ago to characterize the human microbial communities (microbiota) present at specific body sites, and to analyze the role of these microbes in human health and disease. It was expected that more knowledge of our microbiome leads to more effective ways to diagnose, treat and eventually prevent diseases. This paper reviews the skin microbiome research and the relation of microorganisms to skin diseases. © 2014 De Nederlandse Vereniging voor Dermatologie en Venereologie.
Article
Our group recently discovered that certain dietary nutrients possessing a trimethylamine (TMA) moiety, namely choline/phosphatidylcholine and L-carnitine, participate in the development of atherosclerotic heart disease. A meta-organismal pathway was elucidated involving gut microbiota-dependent formation of TMA and host hepatic flavin monooxygenase 3-dependent (FMO3-dependent) formation of TMA-N-oxide (TMAO), a metabolite shown to be both mechanistically linked to atherosclerosis and whose levels are strongly linked to cardiovascular disease (CVD) risks. Collectively, these studies reveal that nutrient precursors, gut microbiota, and host participants along the meta-organismal pathway elucidated may serve as new targets for the prevention and treatment of CVD.
Article
Background and aims: Fecal microbiota transplantation (FMT) has gained interest as a novel treatment option for inflammatory bowel diseases (IBD). While publications describing FMT as therapy for IBD have more than doubled since 2012, research that investigates FMT treatment efficacy has been scarce. We conducted a systematic review and meta-analysis to evaluate the efficacy of FMT as treatment for patients with IBD. Methods: A systematic literature search was performed through May 2014. Inclusion criteria required FMT as the primary therapeutic agent. Clinical remission (CR) and/or mucosal healing were defined as primary outcomes. Studies were excluded if they did not report clinical outcomes or included patients with infections. Results: Eighteen studies (9 cohort studies, 8 case studies and 1 randomized controlled trial) were included. 122 patients were described (79 ulcerative colitis (UC); 39 Crohn's disease (CD); 4 IBD unclassified). Overall, 45% (54/119) of patients achieved CR during follow-up. Among the cohort studies, the pooled proportion of patients that achieved CR was 36.2% (95% CI 17.4%–60.4%), with a moderate risk of heterogeneity (Cochran's Q, P = 0.011; I2 = 37%). Subgroup analyses demonstrated a pooled estimate of clinical remission of 22% (95% CI 10.4%–40.8%) for UC (P = 0.37; I2 = 0%) and 60.5% (95% CI 28.4%–85.6%) for CD (P = 0.05; I2 = 37%). Six studies performed microbiota analysis. Conclusions: This analysis suggests that FMT is a safe, but variably efficacious treatment for IBD. More randomized controlled trials are needed and should investigate frequency of FMT administration, donor selection and standardization of microbiome analysis.