You are on page 1of 8

PERSPECTIVES

MICROBIOME
into question13–16. We would argue that
OPINION
the concept of a pathogen is still valid for
studying communicable diseases, such
The germ-organ theory of as tuberculosis, but it does not provide
a theoretical framework that adequately
non-communicable diseases captures the contribution of microbial
communities to non-communicable diseases
(BOX 2). In other words, rather than omitting
Mariana X. Byndloss and Andreas J. Bäumler the term ‘pathogen’, the core concept of the
germ theory, we should be mindful of using
Abstract | Gut dysbiosis is associated with many non-communicable human it in the context of communicable diseases
diseases, but the mechanisms maintaining homeostasis remain incompletely while independently developing a new
understood. Recent insights suggest that during homeostasis, epithelial hypoxia theory to guide studies on the microbial
limits oxygen availability in the colon, thereby maintaining a balanced microbiota origin of non-communicable diseases. In
that functions as a microbial organ, producing metabolites contributing to host this Opinion article, we discuss the most
recent findings of microbiota research in
nutrition, immune education and niche protection. Dysbiosis is characterized by a search of a useful theoretical framework
shift in the microbial community structure from obligate to facultative anaerobes, that adequately captures the contribution
suggesting oxygen as an important ecological driver of microbial organ of gut-associated microbial communities
dysfunction. The ensuing disruption of gut homeostasis can lead to non- to human non-communicable diseases
communicable disease because microbiota-derived metabolites are either and its implication for guiding future
research efforts.
depleted or generated at harmful concentrations. This Opinion article describes
the concept that host control over the microbial ecosystem in the colon is critical The microbial organ of the large bowel
for the composition and function of our microbial organ, which provides a More than 2 millennia have passed
theoretical framework for linking microorganisms to non-communicable diseases. since Hippocrates (circa 460–370 bce)
commented “all disease begins in the gut”,
but in light of recent data revealing how
The germ theory triggered a scientific and even neurological disorders11,12. microorganisms that inhabit our body
revolution during the time of Robert However, research into the microbial origin affect human health, this quote feels more
Koch (1843–1910) and Louis Pasteur of non-communicable human diseases timely than ever 17. The gut microbiota, the
(1822–1895) by guiding efforts to identify differs in one important aspect from the largest microbial community inhabiting
bacterial pathogens as leading causes studies performed at the end of the 19th our body, fulfils many functions important
of communicable diseases (BOX 1). By century: the identification of pathogens as for human physiology, including nutrition,
developing culture-dependent methods for causes of communicable human diseases development of the immune system and
studying microorganisms, Louis Pasteur was guided by Louis Pasteur’s germ theory, niche protection against enteric pathogens18
laid the foundation for a new discipline, whereas contemporary studies on the role (FIG. 1). The microbiota of the large bowel
bacteriology. Similarly, recent advances in of the gut microbiota in health and disease is of particular importance and forms the
culture-independent methods for analysing lack an overarching theoretical framework focus of this Opinion article because in the
microorganisms have opened up a new for guidance (BOX 2) and therefore colon, bacterial density reaches a staggering
field of study, microbiota research, that remain heavily focused on technological 1011 organisms per gram, making this
parallels the remarkable advances made development, census taking and predicting bacterial community the principal source of
during the ‘golden age’ of microbiology physiological potential. microbial metabolites in the human body.
in the 19th century. Whereas the initial The germ theory influenced initial In turn, these metabolites are responsible for
use of culture-dependent methods by studies on the microbial origin of non- the beneficial effects of the gut microbiota
Louis Pasteur and his contemporaries communicable diseases as investigators on host physiology 18. It was thus proposed
established a microbial cause for many used 16S ribosomal RNA gene profiling in 2005 that the gut-associated microbial
communicable human diseases, recent to identify pathogens linked to an illness, community of the large bowel represents
advances in culture-independent methods which is understandable given the focus a ‘microbial organ’ (REF. 19) because this
have made it possible to associate on the contribution of microorganisms collection of microorganisms resides as a
gut-associated microbial communities with to disease. However, the realization that structural unit and has a common function,
many non-communicable human diseases, most non-communicable diseases are not which is the production of metabolites
including obesity 1,2, arthritis3, asthma4,5, associated with a pathogen resulted in a at concentrations that promote health.
cardiovascular disease6, cancer7–9, diabetes10 conceptual crisis, calling the term ‘pathogen’ Although this idea is generally accepted17,

NATURE REVIEWS | MICROBIOLOGY VOLUME 16 | FEBRUARY 2018 | 103


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

Box 1 | The germ theory of communicable diseases mammalian gut because the diversity of
this microbial community in adults does
Girolamo Fracastoro is credited with proposing in 1546 that microorganisms might cause human not suggest selection for any particular
diseases. However, this concept did not gain acceptance because a connection could not be bacterial species20. There is evidence that a
established until the invention of culture-dependent methods for isolating microorganisms, which mammalian host directs the composition of
was initiated by the development of pasteurization by Louis Pasteur in 1864 (REF. 99). This
the gut microbiota during infancy through
technological advance made it possible to associate microorganisms with communicable disease,
a link first demonstrated for an illness of silkworms by Louis Pasteur in 1870 (REF. 100). The first milk oligosaccharides28 and to some degree
human diseases for which an association with microbial pathogens was proposed were leprosy101 throughout life by epithelial release of
and amoebic dysentery in 1875 (REF. 102), anthrax in 1877 (REF. 103), typhoid in 1880 (REF. 104) and antimicrobials, such as defensins29,30. It is
pneumococcal pneumonia in 1881 (REFS 105,106). However, 1.5 decades after the invention of also well established that anaerobiosis is
pasteurization, evidence for a microbial origin of human diseases remained correlative. Similarly, preserved in the mammalian large bowel, but
1.5 decades after the advent of next-generation sequencing, contemporary microbiota research the idea that this global ecosystem control is
has mostly linked the gut microbiota with human diseases rather than proving causation17. Thus, imposed by the host emerged only recently.
the innovations of culture-dependent and culture-independent methods were followed by an We now know that mammalian hosts restrict
initial phase of intense research that remained largely correlative. oxygen availability in the lumen of the large
In 1882, Robert Koch proposed a game-changing approach ascertaining cause and effect,
bowel, thereby driving the composition
termed ‘Koch’s postulates’, to conclusively establish the microbial origin of communicable disease,
which he first applied to tuberculosis, the leading cause of human mortality at the time107. The of the microbial community towards a
ability to prove causation spawned a flurry of seminal contributions over the following 2 decades, dominance of obligate anaerobes, which is
including the identification of microbial causes for cholera108,109, typhoid110, diphtheria111, critical for maintaining gut homeostasis31.
tetanus112, bubonic plague113,114 and bacillary dysentery115, the identification of the first virulence Epithelial cells are maintained in a state of
factors (diphtheria toxin116 and Shiga toxin117,118) and the development of a treatment for hypoxia (<1% oxygen) at the colonic surface,
diphtheria119,120 and vaccines against anthrax121, rabies122 and typhoid123. The discoveries made which limits the diffusion of oxygen from
during the golden age of microbiology firmly established the germ theory, the idea that a specific the colonic mucosa into the intestinal lumen,
microbial pathogen causes a discrete communicable human disease. This concept set the germ an attribute that preserves anaerobiosis in the
theory apart from competing ideas, which held that microorganisms would be transformed into large bowel32. In turn, anaerobiosis ensures
pathogenic forms through exposure to miasma, a type of poisonous vapour. The identification of
a dominance of obligate anaerobic bacteria
pathogens facilitated vaccine development and made it possible to diagnose infected individuals,
thereby advancing public health measures to curb the spread of disease. It also identified bacterial that belong to the Bacteroidetes phylum and
pathogens as a target for treatment, which eventually led to the development of antibiotics. Clostridia class33. This feature is conserved
between individuals, although microbial
communities tolerate an intimidating
it has never become an alternative to the they are present as quantitatively minor degree of species diversity 20. Constraining
germ theory. One aspect that has curbed components of the gut microbiota25. The oxygen availability is critical because
the use of the microbial organ concept as a quest to identify pathobionts by profiling under anaerobic conditions, our microbial
guide for microbiota research is the daunting microbial communities in health and organ self-assembles into a community of
complexity of our gut-associated microbial disease follows in the footsteps of the microorganisms that stably coexist34 by
community. Comparison of the gut search for pathogens during the golden forming an anaerobic trophic network35. Each
microbiota composition between different age of microbiology (BOX 1), illustrating position in this trophic network represents
individuals reveals very little overlap on the the marked influence that the germ theory a nutrient niche that can be occupied by
species level20, thus making it problematic continues to exert on microbiota research. any microorganism that discharges the
to define what a ‘balanced microbial However, although a focus on individual required metabolic functions. Each occupant
community’ or a functional microbial organ microorganisms is appropriate for studying of a nutrient niche is predicted to be able
should look like. As a result of limited pathogens that cause communicable diseases, to use a few limiting resources better than
progress in understanding how a balanced this viewpoint is too narrow to fully capture any other member within the microbial
microbial community is maintained during the contribution of microbial community community, and the abundance of these
gut homeostasis, the microbial organ ecology to health and disease (BOX 2). Recent limiting resources determines the abundance
concept has long remained elusive, thus insights into how the host controls the gut of the respective occupant, a concept known
curtailing its use as a conceptual framework. ecosystem to ensure homeostasis suggest as the nutrient niche hypothesis36. The
Our poor understanding of gut that it is time to revisit the usefulness of the formation of this anaerobic trophic network
homeostasis has shifted the focus to microbial organ concept as a comprehensive generates metabolite profiles that are similar
aspects that are easier to grasp, such as the guide for microbiota research. among individuals, regardless of the species
association of disease with the presence Invertebrate models reveal a striking composition of the microbial community.
or absence of individual microorganisms. control of microbial community composition During gut homeostasis, our microbial
Specifically, the concept that disease by the host, as species-specific antimicrobial organ hydrolyses and ferments complex
can result from a loss of beneficial peptides are believed to be the selective force dietary carbohydrates (fibre) that escape
commensal bacteria or an overgrowth of behind the different bacterial communities degradation by host enzymes in the
resident bacteria that have the potential associated with closely related species of the upper gastrointestinal tract into smaller
to cause disease, termed ‘pathobionts’, is a cnidarian Hydra26, whereas the light organ of compounds, with the short-chain fatty
commonly used guide for contemporary Euprymna scolopes is associated with a single acids acetate, propionate and butyrate being
microbiota research21–24. The term ‘keystone bacterial species, Aliivibrio fischeri 27. At first the most abundant metabolites produced
species’ has been proposed for pathobionts glance, host control over the composition during gut homeostasis37 (FIG. 1). On the
that instigate inflammation even when of the gut microbiota is less evident in the basis of their main metabolic functions,

104 | FEBRUARY 2018 | VOLUME 16 www.nature.com/nrmicro


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

obligate anaerobic bacteria that degrade The picture emerging from these lumen, which gives rise to the most
complex carbohydrates can be grouped into studies is that during gut homeostasis, consistent and robust ecological pattern
butyrate-producing bacteria, saccharolytic the host controls its microbial organ in the observed during gut dysbiosis, a shift in
bacteria, sulfate-reducing bacteria and large bowel through mechanisms that the microbial community structure from
acetate-producing bacteria (acetogens)38. include limiting oxygen availability in this obligate to facultative anaerobes51,52. An
Saccharolytic bacteria produce hydrogen, ecosystem. The latter provides a benefit by expansion of facultative anaerobic bacteria
which is consumed by hydrogenotrophic curbing the ability of facultative anaerobic of the phylum Proteobacteria has been
microorganisms, including sulfate-reducing microorganisms to compete with the host described in individuals consuming a
bacteria, acetogens and methanogens39. for nutrients and by ensuring the production Western-style diet 53,54, undergoing antibiotic
Obligate anaerobic microorganisms that of microbial fermentation products that therapy 55 or suffering from irritable
belong to these metabolic groupings promote nutrition, immune development bowel syndrome56,57, inflammatory bowel
produce a multitude of metabolites in and gut homeostasis. disease58, metabolic syndrome59, necrotizing
addition to short-chain fatty acids, as enterocolitis60 or colorectal cancer 61. An
has been elegantly reviewed elsewhere40. Microbial organ dysfunction expansion of Proteobacteria is also observed
Importantly, the majority of microbiota- Based on the above model of microbial organ in animal models of chemically induced
derived short-chain fatty acids is absorbed function (FIG. 1), we propose that dysbiosis colitis62,63, genetically induced colitis62,64,
by the host 41 and contributes to host is a state of microbial organ dysfunction, colorectal cancer 61, infection-induced
nutrition42. Evolutionarily, anaerobiosis a condition in which the gut-associated colitis62,65,66 or antibiotic-mediated loss of
could be viewed as a host-driven strategy microbial community becomes a liability colonization resistance67–69. The regularity
to limit the ability of the microbiota to because the host no longer maintains proper with which a disruption of gut homeostasis
compete nutritionally with the host for control over the ecosystem. Microbial organ produces an expansion of Proteobacteria
compounds produced by the microbial dysfunction can produce disease because suggests that this is a microbial signature
fermentation of fibre. In the presence of metabolites are either depleted or produced of gut dysbiosis70. Below, we discuss
oxygen, facultative anaerobic bacteria can at a harmful level21–24. As the gut microbiota recent data supporting the concept that
catabolize fermentation products to carbon can produce many different metabolites increased availability of oxygen or other
dioxide. By limiting oxygen availability, that are capable of acting both locally exogenous electron acceptors constitutes
the host ensures that obligate anaerobes and on cells in distant tissues40, microbial the main ecological driver of expansion
contribute to its own nutrition by producing organ dysfunction can result in metabolite of Proteobacteria.
metabolites through the fermentation imbalances that affect numerous organs, Increased oxygen availability in the
of fibre while at the same time limiting thereby possibly explaining the broad colon can ensue from disruption of
catabolism of fermentation products to spectrum of non-communicable diseases the gut microbiota, which can be triggered
carbon dioxide by facultative anaerobic associated with dysbiosis17. by either antibiotic therapy or intestinal
bacteria. Through this mechanism, the Importantly, disruption of the virtuous inflammation (FIG. 2). Depletion of gut
host maintains control over the microbial cycle that maintains anaerobiosis in the microorganisms during antibiotic therapy
ecosystem without imposing constraints colon31 (FIG. 1) impairs the ability of the host reduces the production of microbiota-
on the species composition of the to limit the flow of oxygen into the gut derived metabolites, as indicated by a
microbial community.
Microbiota-derived short-chain
fatty acids also contribute to immune Box 2 | Communicable diseases versus non-communicable diseases
development 43–48 and gut homeostasis3,31,49,50.
Communicable diseases received their name because they are transmitted from one person or
The latter process requires a continuous
animal to another, either by direct contact, by exposure to bodily discharge or indirectly through
supply of microbiota-derived short-chain fomites or vectors, such as arthropods. Causes of communicable diseases are infectious agents
fatty acids to stimulate the intracellular that fit the classic definition of a microbial pathogen, such as Mycobacterium tuberculosis, the
butyrate sensor peroxisome proliferator- causative agent of tuberculosis; Plasmodium falciparum, which causes malaria; or HIV, an infection
activated receptor‑γ (PPARγ) in colonic that eventually progresses to AIDS. The notion that pathogens are the cause of communicable
epithelial cells31 and to promote maturation diseases is known as the germ theory (BOX 1). At the centre of this concept is the idea that frank
and expansion of regulatory T cells in the pathogens can overcome host defences in individuals with an intact immune system. Thus,
colonic mucosa45–48,50. Regulatory T cells communicability is not due to an underlying defect in the host but is driven by virulence factors of
and epithelial PPARγ signalling cooperate the microbial pathogen. Worldwide, communicable diseases continue to be the leading causes
to drive the metabolism of the colonic of human mortality.
However, in high-income countries, the leading causes of death are non-communicable
surface towards mitochondrial β-oxidation31,
diseases, such as cardiovascular diseases, cancer, chronic respiratory diseases and diabetes.
a process that consumes large quantities Non-communicable diseases were originally defined as medical conditions that are not caused by
of oxygen, thereby rendering epithelial an infectious agent. Surprisingly, research over the past 14 years has linked the resident microbial
cells at the colonic surface hypoxic32. community that inhabits our large bowel to many of those non-communicable human diseases.
Hence, epithelial hypoxia ensures that The germ theory, the leading concept on the microbial origin of human disease, does not apply to
obligate anaerobes produce short-chain non-communicable diseases, because they are not due to infection with communicable infectious
fatty acids, which in turn induce epithelial agents but rather are driven by underlying defects in host physiology that are not communicable.
hypoxia, thereby driving a virtuous cycle Thus, while the germ theory provides a theoretical framework for communicable diseases, there is
that maintains gut homeostasis and a need to develop a new theoretical framework that explains how host physiology balances the
balances the composition of the microbial gut-associated microbial community and how defects in host physiology disrupt this balance,
thereby linking microorganisms to non-communicable human diseases.
community 31 (FIG. 1).

NATURE REVIEWS | MICROBIOLOGY VOLUME 16 | FEBRUARY 2018 | 105


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

Secondary Niche
oxygenation of the colonic surface78 because
Clostridium
bile acids protection difficile this cell type obtains energy through
anaerobic glycolysis76. The consequent
Microbial organ disruption in anaerobiosis drives an
Diet (obligate Metabolites expansion of Proteobacteria in the colon
anaerobes)
through aerobic respiration78. Importantly, as
discussed below, dysbiosis can lead to disease
Butyrate Propionate Acetate by mechanisms that are not linked to the
Mature expansion of Proteobacteria6,81,82. However,
colonocyte Anaerobiosis Nutrition Lumen an expansion of this taxon is relevant because
it indicates an increased bioavailability of
Immune host-derived respiratory electron acceptors,
development which points to an impaired ability of the
host to control the gut ecosystem as an
O2 CO2 important driver of dysbiosis.
Although mild intestinal inflammation
O2 O2 gradient
increases epithelial oxygenation31,74 (FIGS 2,3),
Crypt
severe intestinal inflammation can render
0.1%
the mucosal surface hypoxic. During severe
Blood vessel intestinal inflammation, necrosis of the upper
1% mucosa leads to loss of the epithelial surface
Lactate Regulatory T cells and formation of a pseudomembrane that
Dividing
Glucose
is composed of necrotic tissue, neutrophils
colonocyte 10% and fibrin83. This pseudomembrane becomes
Figure 1 | The host controls the gut ecosystem to maintain homeostasis. Hypoxia of the colonic hypoxic because neutrophils covering the
epithelial cells (colonocytes) lining the mucosal surface maintains luminal anaerobiosis,
Nature which drives
Reviews | Microbiology surface undergo a respiratory burst that
the composition of the gut microbiota towards a dominance of obligate anaerobes. Butyrate produced depletes oxygen84. Although the mucosal
by this microbial organ is oxidized by the colonic epithelium to carbon dioxide (CO2) to preserve surface is hypoxic, these pathological changes
epithelial hypoxia (<1% oxygen (O2)), thereby maintaining gut homeostasis. Dividing colonocytes at drive an expansion of Proteobacteria,
the bottom of crypts obtain energy through anaerobic glycolysis (that is, the fermentation of glucose such as Salmonella enterica65 and Yersinia
to lactate) and are thus not hypoxic. A healthy diet ensures that the microbial organ produces enterocolitica85, as the neutrophil respiratory
metabo­lites that function in nutrition, immune development and niche protection. For simplicity, burst generates electron acceptors for
only examples of metabolites are shown. The O2 concentration is indicated by the red colouring of
anaerobic bacterial respiration, such as
the schematic.
tetrathionate, which promote growth of
these enteric pathogens. Thus, depletion of
marked drop in the concentration of As oxygen originates from the host short-chain fatty acids (FIG. 2), colonic crypt
short-chain fatty acids that are crucial for the epithelium, an expansion of Proteobacteria hyperplasia (FIG. 3) and pseudomembrane
maintenance of gut homeostasis71. Intestinal could be viewed as a microbial signature of formation each drive an expansion of
inflammation induced by enteric pathogens, epithelial dysfunction31. Proteobacteria by increasing the availability
such as Salmonella serovars (phylum A second mechanism that can lead of host-derived exogenous respiratory
Proteobacteria), causes transepithelial to increased oxygen availability in the electron acceptors in the lumen of the
migration of neutrophils that deplete colon is induction of excessive epithelial large bowel.
Clostridia72,73, thereby lowering short-chain repair mechanisms (FIG. 3). Ulcerative It is important to keep in mind that
fatty acid levels in the colon74. The depletion colitis is associated with an expansion although increased availability of respiratory
of short-chain fatty acids by antibiotics of Proteobacteria70 that is indicative of a electron acceptors contributes to the
or host neutrophils shifts the energy disruption in anaerobiosis51. Consistent with expansion of Proteobacteria, it may not
metabolism of colonic epithelial cells from this idea, recent work from animal models be the only driver. For instance, antibiotic
mitochondrial β‑oxidation to anaerobic of ulcerative colitis, including dextran treatment increases the availability
glycolysis31,75. As the latter pathway does not sulfate sodium (DSS)-induced colitis and of monosaccharides, which have an
consume oxygen76, epithelial oxygenation Citrobacter rodentium-induced colitis, important role in the observed expansion
increases, which in turn increases the levels suggests that the mechanism driving of Proteobacteria following antibiotic
of oxygen emanating from the epithelial expansion of Proteobacteria is increased treatment86,87. Furthermore, whereas some
surface. An antibiotic-mediated increase oxygen availability in the colon77,78. Epithelial Proteobacteria are pathogens or pathobionts,
in oxygen availability may explain why damage induced during DSS treatment or most members of this taxon are commensal
treatment of mice with streptomycin C. rodentium infection triggers colonic crypt microorganisms, and the expansion of such
increases the redox potential in the caecum hyperplasia, an excessive epithelial repair commensals does not drive disease but
to levels that approximate an aerobic broth response that results in crypt elongation, does represent a biomarker for dysbiotic
culture71. The consequent disruption a reduction of goblet cell numbers and the ecosystem disruption. In other words,
in anaerobiosis drives an expansion of appearance of undifferentiated epithelial although an increase in the abundance of
facultative anaerobic Proteobacteria cells at the luminal surface79,80. The presence Proteobacteria may be observed during
regardless of their pathogenic potential31,74. of undifferentiated epithelial cells increases dysbiosis, disease might be caused by

106 | FEBRUARY 2018 | VOLUME 16 www.nature.com/nrmicro


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

additional metabolite imbalances that are not theoretical framework to describe the microbial organ, whereas disruption
linked to electron acceptor availability or the microbial organ physiology. During gut of host control triggers microbial organ
presence of Proteobacteria. The exact nature homeostasis, the host controls the microbial dysfunction, which provides a link between
of the metabolite imbalance that develops ecosystem97 by limiting luminal oxygen the gut microbiota and non-communicable
during dysbiosis depends on the factors availability in the colon, an ecological diseases. The focus on host control over
that trigger the disruption of the ecosystem. input that is critical for maintaining the microbial ecosystem sets the concept
For example, the antibiotic-mediated a balanced microbial community 31. This of the germ-organ theory apart from the
disruption of the microbiota drives an balanced microbial community functions germ theory, which pictures the pathogen
expansion of Proteobacteria, but antibiotic- as a microbial organ, producing metabolites in control (BOX 2).
associated colitis is caused by an expansion that maintain a virtuous cycle to preserve
of toxin-producing Clostridium difficile, an epithelial hypoxia (FIG. 1). Microbial organ Conclusions
opportunistic pathogen that is an obligate dysfunction represents an ecosystem Broadening the concepts guiding
anaerobe88. Antibiotic treatment lowers disturbance, commonly involving a shift in microbiota research towards an ecological
colonization resistance against C. difficile by the microbial community from obligate to perspective of microbial organ function
depleting other obligate anaerobic members facultative anaerobes, which is indicative puts the spotlight on questions that are
of the microbiota that are producers of of a defect in the ability of the host to limit currently just ‘wallflowers’ in contemporary
secondary bile acids89,90, which are the flow of oxygen and other exogenous microbiota research. For instance, the
metabolites that inhibit growth of vegetative electron acceptors into the gut lumen. germ-organ theory suggests that non-
C. difficile cells91 (FIG. 1). Restoration This loss of host control disrupts the communicable diseases associated with
of a balanced community of obligate microbial ecosystem, thereby contributing an expansion of Proteobacteria can be
anaerobic bacteria by faecal microbiota to non-communicable diseases because linked to impaired host control over the
transplantation can restore microbial organ a microbiota-derived metabolite is either gut ecosystem. One example of impaired
function and thus provides an effective depleted or produced at harmful levels. We host control is epithelial dysfunction, which
treatment for antibiotic-associated colitis92,93. propose the term ‘germ-organ theory’ to leads to a disruption of anaerobiosis in the
Indeed, inoculation of the secondary bile describe the concept that host control over large bowel31. We do not know whether
acid-producing Clostridium scindens was the microbial ecosystem in the large bowel is depleting short-chain fatty acids (FIG. 2)
shown to restore levels of secondary bile critical for the composition and function of or inducing excessive epithelial repair
acids in antibiotic-treated mice, thereby
re-establishing colonization resistance
against C. difficile81. Antibiotics
Another example is consumption of a
te anaero
Western-style diet, which is associated with iga
be
l

Butyrate
Ob

an expansion of Proteobacteria53,54 but is


s

Disruption
linked to disease because dietary inputs of the
alter the metabolite profile produced by ro microbiota Propionate
t e o b c t e ri
P

the gut microbiota. A Western-style diet a


is associated with obesity, a condition that Acetate
increases the risk of atherosclerosis94. A link Mature
colonocyte O2
between diet and cardiovascular disease is Lumen
the consumption of l‑carnitine and choline,
both of which are abundant in red meat,
a common component of a Western-style O2
diet, and are converted by the gut microbiota Glucose Lactate
to trimethylamine (TMA)6,95. TMA is
O2 gradient
absorbed by the host and oxidized by human Neutrophils
O2
hepatic flavin-containing monooxygenases
Crypt 0.1%
to TMA N‑oxide (TMAO), a metabolite
that increases the risk of atherosclerosis6,82.
Pathways to convert l‑carnitine and choline Blood vessel 1%
into TMA are present in bacteria that Inflammation
belong to various phyla6,95,96. Thus, although Dividing
antibiotic treatment and the consumption colonocyte Regulatory T cell 10%
of a Western-style diet both disrupt gut
homeostasis, as indicated by an expansion Figure 2 | Microbiota disruption causes microbial organ dysfunction. Antibiotic treatment or the
of Proteobacteria, each is linked to disease migration of neutrophils into the intestinal lumen during severe intestinal
Natureinflammation can cause
Reviews | Microbiology
the disruption of the microbiota, which leads to a depletion of short-chain fatty acids, thereby shift-
through a different metabolite imbalance.
ing the metabolism of mature colonic epithelial cells (colonocytes) from mitochondrial β‑oxidation to
anaerobic glycolysis (that is, the fermentation of glucose to lactate). The resulting increase in epithelial
The germ-organ theory oxygenation disrupts anaerobiosis, which drives an expansion of facultative anaerobic Proteobacteria,
Recent findings on the mechanisms that a microbial signature of dysbiosis. Neutrophils that encounter bacteria (for example, during trans­
maintain or disrupt gut homeostasis epithelial migration into the lumen) undergo a respiratory burst and become hypoxic (<1% oxygen
can be assembled into the following (O2))124. The O2 concentration is indicated by the red colouring of the schematic.

NATURE REVIEWS | MICROBIOLOGY VOLUME 16 | FEBRUARY 2018 | 107


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

service facilities, mechanistic follow‑up


studies aimed at understanding trophic
te anaero networks, the influence of host physiology
iga
on the microbial ecosystem and the role

be
l
Ob

s
Metabolites Lumen that microbiota-derived metabolites
have in health and disease will require
ro
t e o b a c t e ri expertise in both bacterial physiology and

a
Epithelial Dividing immunology, which can be obtained only
Butyrate Propionate Acetate
damage colonocyte O2 by attracting scientists from these fields
to enter microbiota research. Researchers
in the field of bacterial pathogenesis
O2
seem particularly well prepared to enter
Lactate this arena as their research exposes them
Glucose
to bacterial physiology as well as to the
interaction of pathogens with the immune
O2 system, thus providing an ideal background
Epithelial for mechanistic microbiota research.
repair O2 gradient
response Alternatively, collaboration between
Crypt
hyperplasia Blood vessel 0.1% scientists engaged in bioinformatics,
bacterial physiology and ecology,
Regulatory T cells cell biology and immunology could
1% accelerate decryption of the mechanistic
basis for microbial contributions to
non-communicable diseases. Although
10%
funding agencies have not provided major
Figure 3 | Excessive epithelial repair triggers microbial organ dysfunction. Epithelial damage incentives for bacterial physiologists
triggers epithelial repair responses, which are characterized by cryptNature Reviews
elongation | Microbiology
in the presence of and immunologists to enter microbiota
dividing colonocytes at the epithelial surface. These immature dividing colonocytes obtain energy research, history suggests that this might
through anaerobic glycolysis (that is, the fermentation of glucose to lactate), which renders the colonic be a move in the right direction as we stand
surface normoxic (3–10% oxygen (O2)). The resulting disruption of anaerobiosis drives an expansion at the threshold of a ‘second golden age
of facultative anaerobic Proteobacteria, a microbial signature of dysbiosis. The O2 concentration is of microbiology’.
indicated by the red colouring of the schematic.
Mariana X. Byndloss and Andreas J. Bäumler are at
the Department of Medical Microbiology and
(FIG. 3) are the only mechanisms increasing niche within our microbial organ or the Immunology, School of Medicine, University of
epithelial oxygenation in the colon. More metabolic functions that make a specific California, Davis, California 95616, USA.
research is needed to unravel the causes microorganism a suitable occupant of a Correspondence to A.J.B.
of epithelial dysfunction for the various particular nutrient niche. Shedding light ajbaumler@ucdavis.edu
non-communicable diseases associated on these crucial knowledge gaps will be doi:10.1038/nrmicro.2017.158
with expansion of Proteobacteria. The necessary to incorporate an improved Published online 8 Jan 2018
focus on changes in host physiology that understanding of trophic networks into the 1. Turnbaugh, P. J. et al. An obesity-associated gut
impair control over the gut ecosystem germ-organ theory, thereby increasing its microbiome with increased capacity for energy
harvest. Nature 444, 1027–1031 (2006).
represents a paradigm shift from the power to predict additional mechanisms 2. Henao-Mejia, J. et al. Inflammasome-mediated
current focus on identifying pathobionts through which the host controls the dysbiosis regulates progression of NAFLD and
obesity. Nature 482, 179–185 (2012).
or microbial biomarkers of disease within ecosystem. Ecosystem drivers in addition 3. Maslowski, K. M. et al. Regulation of inflammatory
the gut microbiota. Identifying causes of to oxygen levels might explain dysbiosis in responses by gut microbiota and chemoattractant
receptor GPR43. Nature 461, 1282–1286 (2009).
epithelial dysfunction is relevant because settings where expansion of Proteobacteria 4. Russell, S. L. et al. Early life antibiotic-driven
the germ-organ theory points to host is not observed, but the identities of such changes in microbiota enhance susceptibility to
allergic asthma. EMBO Rep. 13, 440–447 (2012).
signalling pathways as a potential target for drivers remain unknown. Tackling this 5. Trompette, A. et al. Gut microbiota metabolism of
intervention strategies. Such approaches question will require a ‘new bacteriology’ dietary fiber influences allergic airway disease and
hematopoiesis. Nat. Med. 20, 159–166 (2014).
would be aimed at rebalancing the gut (REF. 98) to conduct mechanistic studies on
6. Wang, Z. et al. Gut flora metabolism of
microbiota by limiting the availability of microbial physiology in its natural context phosphatidylcholine promotes cardiovascular disease.
Nature 472, 57–63 (2011).
exogenous electron acceptors and restoring of a gut-associated microbial community. 7. Castellarin, M. et al. Fusobacterium nucleatum
epithelial hypoxia, which could be used to As we venture forward, it is helpful infection is prevalent in human colorectal carcinoma.
Genome Res. 22, 299–306 (2012).
complement faecal microbiota transfer or to reflect on the skill sets best suited to 8. Kostic, A. D. et al. Genomic analysis identifies
to develop alternative treatment strategies. tackle challenges that lie ahead. The association of Fusobacterium with colorectal
carcinoma. Genome Res. 22, 292–298 (2012).
Additionally, the germ-organ theory germ-organ theory instructs a shift from 9. Yoshimoto, S. et al. Obesity-induced gut microbial
advocates for obtaining a more complete microbial community profiling towards metabolite promotes liver cancer through
senescence secretome. Nature 499, 97–101
understanding of the composition and understanding host-mediated control (2013).
function of our microbial organ. We know of microbial organ ecology. Although 10. Cani, P. D. et al. Changes in gut microbiota control
metabolic endotoxemia-induced inflammation in
embarrassingly little about the identity of the profiling approaches required to high-fat diet-induced obesity and diabetes in mice.
limiting resources that define each nutrient tackle this question can be performed by Diabetes 57, 1470–1481 (2008).

108 | FEBRUARY 2018 | VOLUME 16 www.nature.com/nrmicro


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

11. Hsiao, E. Y. et al. Microbiota modulate behavioral 39. Nakamura, N., Lin, H. C., McSweeney, C. S., 64. Garrett, W. S. et al. Enterobacteriaceae act in concert
and physiological abnormalities associated with Mackie, R. I. & Gaskins, H. R. Mechanisms of microbial with the gut microbiota to induce spontaneous and
neurodevelopmental disorders. Cell 155, 1451–1463 hydrogen disposal in the human colon and implications maternally transmitted colitis. Cell Host Microbe 8,
(2013). for health and disease. Annu. Rev. Food Sci. Technol. 1, 292–300 (2010).
12. Minter, M. R. et al. Antibiotic-induced perturbations in 363–395 (2010). 65. Winter, S. E. et al. Gut inflammation provides a
gut microbial diversity influences neuro-inflammation 40. Nicholson, J. K. et al. Host-gut microbiota metabolic respiratory electron acceptor for Salmonella. Nature
and amyloidosis in a murine model of Alzheimer’s interactions. Science 336, 1262–1267 (2012). 467, 426–429 (2010).
disease. Sci. Rep. 6, 30028 (2016). 41. den Besten, G. et al. The role of short-chain fatty acids 66. Haag, L. M. et al. Intestinal microbiota shifts towards
13. Methot, P. O. & Alizon, S. What is a pathogen? Toward in the interplay between diet, gut microbiota, and host elevated commensal Escherichia coli loads abrogate
a process view of host-parasite interactions. Virulence energy metabolism. J. Lipid Res. 54, 2325–2340 colonization resistance against Campylobacter jejuni
5, 775–785 (2014). (2013). in mice. PLoS ONE 7, e35988 (2012).
14. Casadevall, A. & Pirofski, L. A. Microbiology: Ditch the 42. Velazquez, O. C., Lederer, H. M. & Rombeau, J. L. 67. Bohnhoff, M., Drake, B. L. & Miller, C. P. Effect of
term pathogen. Nature 516, 165–166 (2014). Butyrate and the colonocyte. Production, absorption, streptomycin on susceptibility of intestinal tract to
15. Altmann, D. & Boyton, R. Nomenclature: Replace metabolism, and therapeutic implications. Adv. Exp. experimental Salmonella infection. Proc. Soc. Exp.
‘pathogens’ with ‘perceptogens’. Nature 518, 35 Med. Biol. 427, 123–134 (1997). Biol. Med. 86, 132–137 (1954).
(2015). 43. Millard, A. L. et al. Butyrate affects differentiation, 68. Saito, K. Studies on the habitation of pathogenic
16. Sultana, S., Sarker, S. A. & Brussow, H. What happened maturation and function of human monocyte-derived Escherichia coli in the intestinal tract of mice. I.
to Koch’s postulates in diarrhea? Environ. Microbiol. dendritic cells and macrophages. Clin. Exp. Immunol. Comparative experiments on the habitation of each
http://dx.doi.org/10.1111/1462-2920.13787 (2017). 130, 245–255 (2002). type of resistant pathogenic Escherichia coli under
17. Cani, P. D. Gut microbiota — at the intersection of 44. Wang, B., Morinobu, A., Horiuchi, M., Liu, J. & an administration of streptomycin [Japanese].
everything? Nat. Rev. Gastroenterol. Hepatol. 14, Kumagai, S. Butyrate inhibits functional differentiation Paediatr. Jpn. 65, 385–393 (1961).
321–322 (2017). of human monocyte-derived dendritic cells. 69. Saito, K. Studies on the habitation of pathogenic
18. Lopez, C. A., Kingsbury, D. D., Velazquez, E. M. & Cell. Immunol. 253, 54–58 (2008). Escherichia coli in the intestinal tract of mice. II.
Baumler, A. J. Collateral damage: microbiota-derived 45. Atarashi, K. et al. Induction of colonic regulatory T cells Experimental inoculation of type 055 Escherichia coli
metabolites and immune function in the antibiotic era. by indigenous Clostridium species. Science 331, after long-term administration of streptomycin
Cell Host Microbe 16, 156–163 (2014). 337–341 (2011). [Japanese]. Paediatr. Jpn. 65, 394–399 (1961).
19. O’Hara, A. M. & Shanahan, F. The gut flora as a 46. Arpaia, N. et al. Metabolites produced by commensal 70. Shin, N. R., Whon, T. W. & Bae, J. W. Proteobacteria:
forgotten organ. EMBO Rep. 7, 688–693 (2006). bacteria promote peripheral regulatory T‑cell microbial signature of dysbiosis in gut microbiota.
20. Tap, J. et al. Towards the human intestinal microbiota generation. Nature 504, 451–455 (2013). Trends Biotechnol. 33, 496–503 (2015).
phylogenetic core. Environ. Microbiol. 11, 47. Furusawa, Y. et al. Commensal microbe-derived 71. Meynell, G. G. Antibacterial mechanisms of the
2574–2584 (2009). butyrate induces the differentiation of colonic mouse gut. II. The role of Eh and volatile fatty acids
21. Shreiner, A., Huffnagle, G. B. & Noverr, M. C. regulatory T cells. Nature 504, 446–450 (2013). in the normal gut. Br. J. Exp. Pathol. 44, 209–219
The “Microflora Hypothesis” of allergic disease. 48. Smith, P. M. et al. The microbial metabolites, short- (1963).
Adv. Exp. Med. Biol. 635, 113–134 (2008). chain fatty acids, regulate colonic Treg cell 72. Gill, N. et al. Neutrophil elastase alters the murine gut
22. Packey, C. D. & Sartor, R. B. Commensal bacteria, homeostasis. Science 341, 569–573 (2013). microbiota resulting in enhanced Salmonella
traditional and opportunistic pathogens, dysbiosis 49. Chang, P. V., Hao, L., Offermanns, S. & Medzhitov, R. colonization. PLoS ONE 7, e49646 (2012).
and bacterial killing in inflammatory bowel diseases. The microbial metabolite butyrate regulates intestinal 73. Deatherage Kaiser, B. L. et al. A multi-omic view of
Curr. Opin. Infect. Dis. 22, 292–301 (2009). macrophage function via histone deacetylase host-pathogen-commensal interplay in -mediated
23. Round, J. L. & Mazmanian, S. K. The gut microbiota inhibition. Proc. Natl Acad. Sci. USA 111, 2247–2252 intestinal infection. PLoS ONE 8, e67155 (2013).
shapes intestinal immune responses during health and (2014). 74. Rivera-Chavez, F. et al. Depletion of butyrate-
disease. Nat. Rev. Immunol. 9, 313–323 (2009). 50. Singh, N. et al. Activation of Gpr109a, receptor for producing Clostridia from the gut microbiota drives an
24. Bjorksten, B. in Microbial–Host Interaction: Tolerance niacin and the commensal metabolite butyrate, aerobic luminal expansion of Salmonella. Cell Host
versus Allergy. 64th Nestlé Nutrition Institute suppresses colonic inflammation and carcinogenesis. Microbe 19, 443–454 (2016).
Workshop, Pediatric Program (eds Brandtzaeg, P., Immunity 40, 128–139 (2014). 75. Kelly, C. J. et al. Crosstalk between microbiota-derived
Isolauri, E. & Prescott, S. L.) 11–22 (Sydney, 2008). 51. Rigottier-Gois, L. Dysbiosis in inflammatory bowel short-chain fatty acids and intestinal epithelial HIF
25. Hajishengallis, G., Darveau, R. P. & Curtis, M. A. The diseases: the oxygen hypothesis. ISME J. 7, augments tissue barrier function. Cell Host Microbe
keystone-pathogen hypothesis. Nat. Rev. Microbiol. 1256–1261 (2013). 17, 662–671 (2015).
10, 717–725 (2012). 52. Rivera-Chavez, F., Lopez, C. A. & Baumler, A. J. 76. Fan, Y. Y. et al. A bioassay to measure energy
26. Franzenburg, S. et al. Distinct antimicrobial peptide Oxygen as a driver of gut dysbiosis. Free Radic. Biol. metabolism in mouse colonic crypts, organoids, and
expression determines host species-specific bacterial Med. 105, 93–101 (2017). sorted stem cells. Am. J. Physiol. Gastrointest. Liver
associations. Proc. Natl Acad. Sci. USA 110, 53. Devkota, S. et al. Dietary-fat-induced taurocholic acid Physiol. 309, G1–G9 (2015).
E3730–E3738 (2013). promotes pathobiont expansion and colitis in Il10−/− 77. Hughes, E. R. et al. Microbial respiration and formate
27. Ruby, E. G. Lessons from a cooperative, bacterial- mice. Nature 487, 104–108 (2012). oxidation as metabolic signatures of inflammation-
animal association: the Vibrio fischeri-Euprymna 54. Martinez-Medina, M. et al. Western diet induces associated dysbiosis. Cell Host Microbe 21, 208–219
scolopes light organ symbiosis. Annu. Rev. Microbiol. dysbiosis with increased E coli in CEABAC10 mice, (2017).
50, 591–624 (1996). alters host barrier function favouring AIEC 78. Lopez, C. A. et al. Virulence factors enhance
28. Garrido, D., Dallas, D. C. & Mills, D. A. Consumption colonisation. Gut 63, 116–124 (2014). Citrobacter rodentium expansion through aerobic
of human milk glycoconjugates by infant-associated 55. Vollaard, E. J., Clasener, H. A. & Janssen, A. J. respiration. Science 353, 1249–1253 (2016).
bifidobacteria: mechanisms and implications. Co‑trimoxazole impairs colonization resistance in 79. Ahmed, I. et al. Critical roles of Notch and Wnt/β-
Microbiology 159, 649–664 (2013). healthy volunteers. J. Antimicrob. Chemother. 30, catenin pathways in the regulation of hyperplasia
29. Salzman, N. H. et al. Enteric defensins are essential 685–691 (1992). and/or colitis in response to bacterial infection. Infect.
regulators of intestinal microbial ecology. Nat. Immunol. 56. Carroll, I. M., Ringel-Kulka, T., Siddle, J. P. & Ringel, Y. Immun. 80, 3107–3121 (2012).
11, 76–83 (2010). Alterations in composition and diversity of the 80. Chandrakesan, P. et al. Novel changes in NF-κB
30. Hayase, E. et al. R-Spondin1 expands Paneth cells and intestinal microbiota in patients with diarrhea- activity during progression and regression phases of
prevents dysbiosis induced by graft-versus-host predominant irritable bowel syndrome. hyperplasia: role of MEK, ERK, and p38. J. Biol. Chem.
disease. J. Exp. Med. http://dx.doi.org/10.1084/ Neurogastroenterol. Motil. 24, 521–530 (2012). 285, 33485–33498 (2010).
jem.20170418 (2017). 57. Krogius-Kurikka, L. et al. Microbial community analysis 81. Buffie, C. G. et al. Precision microbiome reconstitution
31. Byndloss, M. X. et al. Microbiota-activated PPAR‑γ- reveals high level phylogenetic alterations in the overall restores bile acid mediated resistance to Clostridium
signaling inhibits dysbiotic Enterobacteriaceae gastrointestinal microbiota of diarrhoea-predominant difficile. Nature 517, 205–208 (2015).
expansion. Science 357, 570–575 (2017). irritable bowel syndrome sufferers. BMC Gastroenterol. 82. Bennett, B. J. et al. Trimethylamine-N‑oxide, a
32. Furuta, G. T. et al. Hypoxia-inducible factor 9, 95 (2009). metabolite associated with atherosclerosis, exhibits
1‑dependent induction of intestinal trefoil factor 58. Morgan, X. C. et al. Dysfunction of the intestinal complex genetic and dietary regulation. Cell Metab.
protects barrier function during hypoxia. J. Exp. Med. microbiome in inflammatory bowel disease and 17, 49–60 (2013).
193, 1027–1034 (2001). treatment. Genome Biol. 13, R79 (2012). 83. Tsolis, R. M., Adams, L. G., Ficht, T. A. & Baumler, A. J.
33. Eckburg, P. B. et al. Diversity of the human intestinal 59. Ferreira, R. B. et al. The intestinal microbiota plays a Contribution of Salmonella typhimurium virulence
microbial flora. Science 308, 1635–1638 (2005). role in Salmonella-induced colitis independent of factors to diarrheal disease in calves. Infect. Immun.
34. Faith, J. J. et al. The long-term stability of the human pathogen colonization. PLoS ONE 6, e20338 (2011). 67, 4879–4885 (1999).
gut microbiota. Science 341, 1237439 (2013). 60. Normann, E., Fahlen, A., Engstrand, L. & Lilja, H. E. 84. Campbell, E. L. et al. Transmigrating neutrophils shape
35. Rivera-Chavez, F. & Baumler, A. J. The pyromaniac Intestinal microbial profiles in extremely preterm the mucosal microenvironment through localized
inside you: Salmonella metabolism in the host gut. infants with and without necrotizing enterocolitis. oxygen depletion to influence resolution of
Annu. Rev. Microbiol. 69, 31–48 (2015). Acta Paediatr. 102, 129–136 (2013). inflammation. Immunity 40, 66–77 (2014).
36. Freter, R., Brickner, H., Fekete, J., Vickerman, M. M. 61. Arthur, J. C. et al. Intestinal inflammation targets 85. Kamdar, K. et al. Genetic and metabolic signals during
& Carey, K. E. Survival and implantation of Escherichia cancer-inducing activity of the microbiota. Science acute enteric bacterial infection alter the microbiota
coli in the intestinal tract. Infect. Immun. 39, 338, 120–123 (2012). and drive progression to chronic inflammatory disease.
686–703 (1983). 62. Lupp, C. et al. Host-mediated inflammation disrupts Cell Host Microbe 19, 21–31 (2016).
37. Roy, C. C., Kien, C. L., Bouthillier, L. & Levy, E. the intestinal microbiota and promotes the overgrowth 86. Ng, K. M. et al. Microbiota-liberated host sugars
Short-chain fatty acids: ready for prime time? Nutr. Clin. of Enterobacteriaceae. Cell Host Microbe 2, 204 facilitate post-antibiotic expansion of enteric
Pract. 21, 351–366 (2006). (2007). pathogens. Nature 502, 96–99 (2013).
38. Fischbach, M. A. & Sonnenburg, J. L. Eating for two: 63. Winter, S. E. et al. Host-derived nitrate boosts growth 87. Faber, F. et al. Host-mediated sugar oxidation
how metabolism establishes interspecies interactions of E. coli in the inflamed gut. Science 339, 708–711 promotes post-antibiotic pathogen expansion. Nature
in the gut. Cell Host Microbe 10, 336–347 (2011). (2013). 534, 697–699 (2016).

NATURE REVIEWS | MICROBIOLOGY VOLUME 16 | FEBRUARY 2018 | 109


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PERSPECTIVES

88. Kelly, C. P., Pothoulakis, C. & LaMont, J. T. Clostridium 101. Hansen, G. H. A. On the etiology of leprosy. 117. Conradi, H. Über lösliche, durch aseptische autolyse
difficile colitis. N. Engl. J. Med. 330, 257–262 Br. Foreign Med. Chir. Rev. 55, 459–489 (1875). erhaltene giftstoffe von ruhr- und typhus-bazillen.
(1994). 102. Losch, F. Massenhafte entwickelung von amöben im Dtsch. Med. Wochenschr. 29, 26–28 (1903).
89. Sorg, J. A. & Sonenshein, A. L. Bile salts and glycine dickdarm. Arch. Pathol. Anat. Physiol. 65, 196–211 118. Neisser, M. & Shiga, K. Ueber freie receptoren
as cogerminants for Clostridium difficile spores. (1875). von typhus- und dysenteriebazillen und über das
J. Bacteriol. 190, 2505–2512 (2008). 103. Koch, R. Untersuchungen über Bakterien, V. dysenterietoxin. Dtsch. Med. Wochenschr. 29, 61–62
90. Theriot, C. M. et al. Antibiotic-induced shifts in the Die aetiologie der milzbrandkrankheit, begründet (1903).
mouse gut microbiome and metabolome increase auf der entwicklungsgeschichte des Bacillus 119. von Behring, E. & Kitasato, S. Ueber das
susceptibility to Clostridium difficile infection. anthracis. Beiträge Biol. Pflanzen 2, 277–308 zustandekommen der diphtherie — immunität und der
Nat. Commun. 5, 3114 (2014). (1877). tetanus — immunität bei tieren. Dt. Med. Wochenschr.
91. Wilson, K. H. Efficiency of various bile salt 104. Eberth, K. J. Die organismen in den organen bei 16, 1113–1114 (1890).
preparations for stimulation of Clostridium difficile typhus abdominalis [German]. Virchows Arch. 81, 120. von Behring, E. Untersuchungen über das
spore germination. J. Clin. Microbiol. 18, 1017–1019 58–74 (1880). zustandekommen de diphtherie — immunität bei
(1983). 105. Pasteur, L. Note sur la maladie nouvelle provoquee par tieren. Dt. Med. Wochenschr. 16, 1145–1148
92. van Nood, E. et al. Duodenal infusion of donor feces la salive d’un enfant mort de la rage [French]. (1890).
for recurrent Clostridium difficile. N. Engl. J. Med. C. R. Acad. Sci. 92, 159–165 (1881). 121. Pasteur, L., Roux, E. & Chamberland, C. Summary
368, 407–415 (2013). 106. Sternberg, G. M. A fatal form of septicaemia, produced report of the experiments conducted at Pouilly-le‑Fort,
93. Weingarden, A. R. et al. Microbiota transplantation by the injection of human saliva. An experimental near Melun, on the anthrax vaccination, 1881.
restores normal fecal bile acid composition in research. Bull. Nat. Board Health USA 2, 781–783 C. R. Acad. Sci. 92, 1378–1383 (1881).
recurrent Clostridium difficile infection. (1881). 122. Pasteur, L. Methode pour prevenir l rage apres
Am. J. Physiol. Gastrointest. Liver Physiol. 306, 107. Koch, R. Die aetiologie der tuberkulose. morsure. C. R. Acad. Sci. 101, 765–772 (1885).
G310–G319 (2014). Berliner Klinische Wochenschrift 15, 221–230 123. Pfeiffer, R. & Kolle, W. Experimentele untersuchungen
94. Fung, T. T. et al. Association between dietary (1882). zur frage der schutzimpfungen des menschen gegen
patterns and plasma biomarkers of obesity and 108. Koch, R. Der zweite bericht der deutschen den typhus abdominalis. Dtsch. Med. Wochenschr.
cardiovascular disease risk. Am. J. Clin. Nutr. 73, cholerakommission. Dt. Med. Wochenschr. 9, 22, 735–737 (1896).
61–67 (2001). 743–744 (1883). 124. Jennewein, J. et al. Low-oxygen tensions found in
95. Zhu, Y. et al. Carnitine metabolism to trimethylamine 109. Koch, R. Ueber die cholerabakterien. Dt. Med. Salmonella-infected gut tissue boost Salmonella
by an unusual Rieske-type oxygenase from human Wochenschr. 10, 725–728 (1884). replication in macrophages by impairing antimicrobial
microbiota. Proc. Natl Acad. Sci. USA 111, 110. Gaffky, G. Zur ätiologie des abdominaltyphus. activity and augmenting Salmonella virulence.
4268–4273 (2014). Mitteilungen Kaiserlichen Gesundheitsamt 2, Cell. Microbiol. 17, 1833–1847 (2015).
96. Rath, S., Heidrich, B., Pieper, D. H. & Vital, M. 372–420 (1884).
Uncovering the trimethylamine-producing bacteria 111. Loeffler, F. Untersuchungen über die bedeutung der Acknowledgements
of the human gut microbiota. Microbiome 5, 54 mikroorganismen für die entstehung der diphtherie Work in the authors’ laboratory was supported by public
(2017). beim menschen, bei der taube und beim kalbe. health service grants AI112445, AI112949 and AI096528
97. Foster, K. R., Schluter, J., Coyte, K. Z. & Rakoff- Mitth. a. d. Kaiserl. Gesundheitsamtes 2, 421–499 (A.J.B.).
Nahoum, S. The evolution of the host microbiome (1884).
as an ecosystem on a leash. Nature 548, 43–51 112. Nicolaier, A. Ueber infectiösen tetanus. Dt. Med. Author contributions
(2017). Wochenschr. 10, 842–844 (1884). M.X.B. and A.J.B. substantially contributed to the discussion
98. Pedron, T., Nigro, G. & Sansonetti, P. J. From 113. Yersin, A. La peste bubonique à Hong-Kong. of content and review and editing of the manuscript before
homeostasis to pathology: decrypting microbe-host Ann. l’Institut Pasteur 8, 662–667 (1894). submission.
symbiotic signals in the intestinal crypt. Philos. Trans. 114. Kitasato, S. The bacillus of bubonic plague. Lancet 144,
R. Soc. Lond. B. Biol. Sci. http://dx.doi.org/10.1098/ 428–430 (1894). Competing interests statement
rstb.2015.0500 (2016). 115. Shiga, K. Ueber den dysenterie-bacillus (Bacillus The authors declare no competing interests.
99. Bordenave, G. Louis Pasteur (1822–1895). Microbes dysenteriae). Zentralbl Bakteriol Orig 24, 913–918
Infect. 5, 553–560 (2003). (1898). Publisher’s note
100. Pasteur, L. Etudes Sur La Maladie Des Vers A Soie. 116. Roux, E., Y. A. Contribution à l’étude de la diphtérie Springer Nature remains neutral with regard to jurisdictional
Vol. 1,2 (Gauthier-Villars, 1870). Ann. Inst. Pasteur 2, 421–499 (1888). claims in published maps and institutional affiliations.

110 | FEBRUARY 2018 | VOLUME 16 www.nature.com/nrmicro


©
2
0
1
8
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like