You are on page 1of 17

REVIEWS

The mucosal immune system:


master regulator of bidirectional
gut–brain communications
Nick Powell1, Marjorie M. Walker2 and Nicholas J. Talley2
Abstract | Communication between the brain and gut is not one-way, but a bidirectional highway
whereby reciprocal signals between the two organ systems are exchanged to coordinate
function. The messengers of this complex dialogue include neural, metabolic, endocrine
and immune mediators responsive to diverse environmental cues, including nutrients and
components of the intestinal microbiota (microbiota–gut–brain axis). We are now starting to
understand how perturbation of these systems affects transition between health and disease.
The pathological repercussions of disordered gut–brain dialogue are probably especially
pertinent in functional gastrointestinal diseases, including IBS and functional dyspepsia.
New insights into these pathways might lead to novel treatment strategies in these common
gastrointestinal diseases. In this Review, we consider the role of the immune system as the
gatekeeper and master regulator of brain–gut and gut–brain communications. Although adaptive
immunity (T cells in particular) participates in this process, there is an emerging role for cells of
the innate immune compartment (including innate lymphoid cells and cells of the mononuclear
phagocyte system). We will also consider how these key immune cells interact with the specific
components of the enteric and central nervous systems, and rapidly respond to environmental
variables, including the microbiota, to alter gut homeostasis.

Globally, gastrointestinal diseases are major causes of gastrointestinal diseases such as IBD, coeliac disease
mortality and morbidity. Infectious diarrhoea remains a and FGID5–7, environ­mental vari­ables are domin­ant and
major cause of death in the developing world, but chronic shape the development and clinical course of these dis-
gastrointestinal diseases are common ­diseases globally. eases. Furthermore, the digestive tract does not operate
Chronic intestinal inflammatory diseases, including independently, but instead requires functional integra-
Crohn’s disease and ulcerative colitis, the two major tion with other organ systems. Crosstalk between the
manifesta­tions of IBD, affect nearly 1% of Western gastrointestinal tract and the ­central nervous system
­societies and the incidence of IBD continues to rise, (CNS) and enteric nervous system (ENS) are impor-
including increases in the number of newly reported cases tant examples of how signals originating in one of these
in the developing world1. However, a considerable burden organ systems affect the function of the other. Of course,
1
Experimental of disease is accounted for by functional gastro­intestinal ­dialogue between these systems has been appreciated for
Immunobiology, Division of diseases (FGIDs), which are defined by character­istic many years. Since the pioneering work of Ivan Petrovich
Transplant Immunology and
Mucosal Biology, Great Maze
patterns of symptoms in the absence of c­ lassic organic Pavlov at the turn of the nineteenth century it has been
Pond, King’s College London, pathology. FGIDs include IBS and functional dys- unequivocally demonstrated that the brain directly influ-
SE1 9RT, UK. pepsia (FD), which are among the most c­ ommonly ences gut function8. By forming fistulae in digestive tract
2
University of Newcastle, ­encountered symptom complexes in most adult popula- organs, including the salivary glands, stomach, duo­
Faculty of Health and
tions. For instance, in Western societies, over one-third denum and pancreas, Pavlov showed that gastro­intestinal
Medicine, School of Medicine
& Public Health, Callaghan of the population has chronic gastrointestinal disorders, secretions could be triggered in response to auditory,
NSW 2308, Australia. dominated by IBS and FD2–4. Accordingly, a pressing visual or painful stimuli (conditional reflexes), and by
Correspondence to N.P. need now exists to understand the patho­physiology of ligating key peripheral nerves he confirmed neural con-
nick.powell@kcl.ac.uk chronic gastrointestinal diseases and the key factors that trol of digestive function. However, it is increasingly
doi:10.1038/nrgastro.2016.191 influence their clinical course to improve patient out- recognized that brain–gut communication pathways are
Published online 18 Jan 2017 comes. Although genetic factors have a role in chronic ­bidirectional. Indeed, signals originating in the gut can

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
7
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
.
REVIEWS

Key points and particularly in the developed world, there has been
a sharp decline in the prevalence of infectious diseases
• Common gastrointestinal diseases, such as IBS, functional dyspepsia and IBD, and a simultaneous increase of chronic inflammatory
are closely linked to psychological morbidity diseases17. Accordingly, benefits and costs of sickness
• This link is driven in part through bidirectional signalling between the brain and gut, behaviour might have shifted. Indeed, unresolved
which reciprocally regulate each other sickness behavioural responses driven by prolonged,
• Growing evidence implicates the importance of immune activation, which might intractable gastrointestinal diseases such as IBD or FGID
be overt (IBD) or more subtle (IBS, functional dyspepsia) in pathological gut–brain might predispose to psychiatric manifestations, chiefly
interactions in the form of depression18,19.
• The composition of the intestinal microbiota affects behaviour and mood, Chronic gut inflammation is linked to a consider-
which could in part rely on selective activation of distinct host cytokine responses able burden of psychological morbidity. Patients with
• Therapeutic targeting of gut microorganisms, host immunity or psychological IBD have increased anxiety and depression20–24, which
symptoms could hold the key to uncoupling pathological interactions between is indepen­dently linked to worse outcomes, includ-
the gut and brain
ing increased disease activity 25,26, increased relapse
rate27,28, reduced response to biological therapy 26 and
poor adherence to treatment 29. Experiments in rodents
also influence brain function, through endocrine, meta­ have provided some important insights on the effect of
bolic, immune and neuronal mediators, which interface psycho­social stress on gut physiology and visceral pain
with the ENS and the CNS9. The ENS is a complex net- modulation, as well as how intestinal inflammation
work of >1 × 108 neurons that can function independently influences these responses. Rodent models have proven
of the CNS, controlling gut motility, endocrine function, extremely valu­able in exploring mechanistic pathways
trans­mucosal fluid movement and local blood flow10. The in these processes, and there are well established and/or
ENS also directly communicates with cells of the intesti- validated models of visceral pain (including colorectal
nal barrier, including neuroendocrine cells and mucosal distension), visceromotor response monitoring, behav-
immune cells11. This neural network is composed of ioural assays and functional imaging 30. Experimental
enteric glia, neurons of peripheral ganglia and epithelial stress induction (including acoustic stress, restraint stress,
sensors such as enteroendocrine cells (EECs), which have water avoidance, maternal deprivation and p ­ redator con-
been termed the gut connectome12. In ­normal digestion, frontation) have demonstrated robustly how stressful
nutrients such as amino acids, peptides and glucose stimuli modulate visceral sensitivity, intestinal motility,
stimulate EECs to produce hormones, which modulate secretory function, intestinal permeability and vulner-
gut motility, hormone prod­uction (for example, insulin ability to intestinal inflammation31–38. Stress induction
secretion), satiety and meta­bolic ­status13,14. Although the in mice is associated with increased susceptibility for
ENS can function as an autono­mous unit (especially in experimental intestinal inflammation (or threshold for
the small and large ­intestine), it is also closely integrated re‑­induction) in different disease models, including dex-
with, and reactive to, the CNS. Important anatomical tran sulfate sodium colitis, DNBS (2,4‑­dinitrobenzene
considerations of the ENS and its interaction with the sulfuric acid) colitis and the spontaneous enterocolitis in
CNS are reviewed in BOX 1. Il10−/− mice. Possible mechanisms include increased gut
In this Review, we will consider the triggers, medi­ leakiness (thereby exposing the mucosal immune s­ ystem
ators and consequences of bidirectional communication to luminal microorganisms), impaired tissue restitu­
between the gut and brain, and how these pathways tion and direct activation of intestinal ­immunity 35–38.
reciprocally affect organ physiology to shape patho­ Inflammation has long-lasting effects on visceral hyper-
logy. Emphasis will be directed at pathways regulated sensitivity with the potential to influence symptoms
by ­normal and aberrant host immune responses to long term. Even after recovery of acute, experimentally
­exogenous stimuli, including the intestinal microbiota. induced colitis (including full mucosal and histological
resolution) there is prolonged alteration in visceral sen-
Gastrointestinal disease and the brain sitivity with reduced threshold for induction of viscero­
Gastrointestinal disease is linked to disordered brain motor responses after colonic distension39. Increased
function. The burden of psychiatric disturbance in intestinal permeability and heightened susceptibility to
patients with gastrointestinal disease is substantial. experimental intestinal inflammation observed in mice
At one of end of the spectrum, chronic inflammation with depressive behaviour (induced by maternal separ­
and cancer in the gut is linked to so‑called sickness ation) is reversed by antidepressant therapy 37. Taken
behaviour, a characteristic behavioural response to together, these important preclinical data support the
illness that is characterized by lethargy, social with- concept that anxiety and depression directly influence
drawal, reduced feeding and reduced sexual activity 15,16. disease biology, not merely gastrointestinal symptoms.
From an evolutionary perspective, sickness behaviour FGIDs are also strongly linked to increased psycho­
has been speculated to have conferred a survival benefit logical morbidity, including anxiety, depression,
in the setting of acute infection by preserving energy, post-traumatic stress disorder and cognitive impair-
avoiding dangerous encounters and limiting the spread ment 40–44, as well as other ‘functional’ comorbidities,
of infection to other community members16. At a popu- including chronic pelvic pain, interstitial cystitis, chronic
lation level, host response to infection has always been fatigue syndrome and fibromyalgia45. Patients with IBS
a key arbiter of survival; however, in the modern era, also exhibit evidence of cognitive deficit, including

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

impairment of visuospatial memory performance, which linked to increased psychological morbidity, the direc-
is independent of psychological comorbidity 46. In com- tion of causality is uncertain. One possibility is that
parison with healthy individuals as controls, patients with psychological disturbance results from chronic and
IBS are more likely to be prescribed with ­antidepressants, debilitating gastrointestinal symptoms, reminiscent of
anxiolytics, antipsychotics and sedatives47. sickness behaviour. Another possibility is that FGIDs
Novel brain imaging approaches, including func- might resemble somatiza­tion disorders, resulting from
tional MRI (fMRI) scanning is now starting to prov­ primary problems with mood, abnormal emotional
ide important new insights into neural activity and responses and a dysregulated perception of pain, which
circuitry in FGIDs. fMRI in patients with IBS demon- manifests with disturbed gut function. Growing evi-
strates that anxiety and depression are associated with dence supports the possibility that both scenarios exist
disturbed patterns in intrinsic brain function, including and that different FGID phenotypes could exist, with
decreased activity in the prefrontal cortex, posterior some patients presenting first with mood disorders
cingulate ­cortex and bilateral inferior parietal cortices48. (without gut symptoms) and then later developing gut
fMRI studies indicate that cognitive processing of vis- symptoms (brain driving gut), whereas others first pres-
ceral pain, anticipation of pain and pain-related fear is ent with FGIDs and then emerge with psycho­logical dis­
defective in patients with IBS, and is typically linked to turbance (gut driving brain). Convincing data to support
excessive engagement of emotional brain networks49–51. this notion have only just emerged, made possible by
study­ing well-defined study participants in longitudinal
FGIDs are bidirectionally associated with psycho- series. In a landmark study, Koloski et al.41 examined a
logical morbidity. A strong association exists between population of 1,775 randomly selected Australian adults
FGIDs and psychological morbidity, and the prevalence who completed a validated questionnaire evaluating
of psychiatric symptoms. Anxiety disorders, depression gastro­intestinal symptoms and symptoms of anxiety
and somatization disorders are ~50% in these patient or depression (the Delusion Symptom States Inventory
groups40,52,53. Early-life stressors, including a history of tool). Patients were recruited in 1997 and a follow‑up
sexual, emotional and physical abuse, are also present survey was conducted 12 years later. The key finding
in 26–44% of patients with FGIDs, and is especially was a clear population of patients with mood disorders
over-represented in patients presenting to second- (­anxiety or depression) at inception, who were ­initially
ary care clinics54–56. Although FGIDs are undoubtedly free of FGID, but during follow‑up subsequently devel-
oped FGIDs. Conversely, individuals with FGIDs with-
out anxiety or depression at baseline had a markedly
Box 1 | The central and enteric nervous systems increased chance of developing anxiety or depression
12 years later 41. The study concluded that the gut was
• Innervation of the gastrointestinal tract regulates secretions, sphincter control,
motility, blood flow and enteroendocrine function probably the driver of psychological morbidity in patients
in whom gastro­intestinal symptoms predated mood dis-
• The enteric nervous system (ENS) is an autonomous neural network with a
comparable number of neurons to the spinal cord, and chiefly comprises myenteric
order, and the brain was probably the driver of FGIDs
ganglia (between the longitudinal and circular muscle layers of the gut wall) in patients in whom anxiety or depression symptoms
and submucosal ganglia manifested before gastrointestinal symptoms. In an
• The myenteric plexus circumferentially encases the gut from the upper oesophagus independent 1‑year prospective follow‑up study of 1,900
to the internal anal sphincter, whereas the submucosal plexus is predominantly people, one-third of individuals had a mood disorder
present in the small and large intestine that preceded FGID symptoms, whereas in two-thirds a
• The three main types of neuron in the ENS are primary sensory neurons, interneurons FGID preceded the mood dis­order 57. Although recogni-
and primary motor neurons; these neurons directly synapse with each other to tion of the directionality of this association might imply
mediate ENS intrinsic reflexes the existence of distinct mechanisms of disease, it is also
• Although the small and large intestine are the key sites of autonomous ENS circuitry, possible that these different modes of presentation (that
it is important to appreciate that the ENS is closely integrated and in extensive is, brain driving gut, or gut driving brain) might instead
communication with the central nervous system (CNS) represent polar ends of the same disease process. A con-
• The main lines of communication between the ENS and CNS are the vagus nerve dition manifests first in one organ system rather than the
(proximal gastrointestinal tract) and the spinal nerves (including thoracolumbar and other by chance.
lumbrosacral networks, which innervate the distal small bowel and colon), all of which Further support for the possibility that psychiatric
contain both sensory (afferent) and motor (efferent) pathways disturbance itself predisposes to the subsequent devel-
• Vagal afferents detecting nutrients, chemicals or luminal contents (chemoreceptors), opment of FGIDs comes from a cohort of patients with
and distention or movement (mechanoreceptors or tension receptors) project to the post-infectious IBS (PI‑IBS) who have been longitu-
nucleus tractus solitarius (NTS) via the dorsal and ventral vagal trunks dinally followed in Walkerton, a rural community in
• Vagal efferents originating in the NTS and nucleus ambiguous directly innervate Canada. PI‑IBS occured in ~10% of patients after an
striated muscle (e.g. oesophagus), but mostly synapse with enteric nerves to mediate acute episode of gastrointestinal infection, even after
motor functions (e.g. motility, secretions, sphincter relaxation) clearance of the offending pathogen58. Heavy rainfall
• The thoracolumbar spinal nerves are composed of sensory neurons with cell bodies in in Walkerton in 2000 resulted in contamination of the
the dorsal root ganglia and efferent sympathetic fibres, which innervate blood vessels regional water source with faeces from grazing live-
and synapse with myenteric and submucosal ganglia
stock, which resulted in a large outbreak of acute gastro­
• The pelvic spinal nerves innervate the rectum and distal colon, and include sensory enteritis predomin­antly with Campylobacter jejuni
(including pain fibres) and motor pathways
and/or Escherichia coli O157:H7. The outbreak affected

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
7
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
.
REVIEWS

>2,000 local residents59. Walkerton residents, includ- Role of the immune system: two-way signals
ing those affected by acute gastroenteritis, and other Building on many decades of research, important
­unaffected individuals were enrolled in a longitudinal advances continue to be made in our understanding of
study to investigate long-term health outcomes in this common pathways responsible for reciprocal modula­
cohort. In comparison with unaffected residents, individ­ tion of function in the brain and gut. Afferent and effer-
uals who developed acute infectious gastroenteritis were ent nerves and blood-borne humoral factors have the
subsequently more likely to develop functional gastro- potential to convey signals between these two organ
intestinal symptoms in long-term follow‑up, includ- systems and, accordingly, factors that regulate these
ing IBS59 and FD60. Importantly, at least in the case of pathways have the potential to impact on gut–brain
post-­infectious FD, gut symptoms were markedly more dialogue. The immune system integrates and modulates
likely to develop in patients with pre-­existing anxiety or bidirectional signals between the brain and the nervous
depression60, consistent with the possibility that mood system and the gut, and consequently mechanistically
disorders predispose to the development of chronic links alterations in function in both brain and gut. Many
gut symptoms. These data are corroborated by other gastrointestinal diseases are linked to inflammation.
indepen­dent cohorts of patients in whom IBS symp- In IBD, these changes are macroscopically evident, but
toms developed following acute bacterial gastro­enteritis, increasingly FGIDs are also being linked to subtle alter-
whereby PI‑IBS was more likely to develop in individuals ations in mucosal immunity. Consequently, the concep-
with pre-­existing psychological morbidity and fatigue61,62. tually attractive possibility exists that aberrant mucosal
However, some of this increased risk is explained by the immune activation directly regulates the effects of stress
increased susceptibility of individuals with pre-existing and anxiety perception in the brain yet, at the same
psychological ­disturbance to infectious gastroenteritis61. time, stress responses originating in the higher cortical
centres have the potential to profoundly affect immune
system activity in the mucosal tissues and directly drive
Box 2 | The mucosal immune system63 gastrointestinal symptoms. To put these concepts into
context it is necessary to understand the dynamics of the
• CD4+ effector T‑cell subsets are defined by the expression of specific transcription
factors and by the pattern of cytokines produced. Type 1 T helper (TH1) cells produce stress response and factors that modulate it (including
IFNγ and TNF, which have many pro-inflammatory actions, including activation of the immune system), but also to consider the immune
macrophages. Type 2 T helper (TH2) cells produce IL‑4, IL‑5 and IL‑13, which promote mechanisms operational in common gastrointestinal
activation, survival and maintenance of tissue mast cells and eosinophils; the TH2 diseases, including FGIDs.
response is implicated in host immunity to multicellular pathogens, such as helminths.
Type 17 T helper (TH17) cells produce IL‑17A, IL‑17F and IL‑22, which support the Mucosal immune system, ENS and stress
recruitment and activation of neutrophils and have an important role in host Reciprocal interactions between mucosal immune
resistance to extracellular bacteria and fungi. Regulatory T cells expressing the system, ENS and stress response. Even under homeo-
transcription factor FOXP3 serve to counterbalance overly exuberant effector
static conditions the gut is described to exist in a state
responses and dampen down inflammation; they secrete immunosuppressive
of ‘physio­logical inflammation’ as, in contrast to other
cytokines, such as IL‑10 and TGFβ, and are one of the main immunoregulatory
mechanisms in the gut (and beyond). tissues, there is relative expansion of infiltrating lympho-
cytes in the lamina propria and intraepithelial compart-
• Key cells of the mucosal innate immune compartment include cells of the mononuclear
phagocyte system (MPS), such as monocytes, macrophages and dendritic cells. ments of both the large and small intestine. These cells are
Intestinal macrophages, which are continually replenished by circulating monocytes, charged with the challenging task of remaining immuno-
are mostly anti-inflammatory cells in the steady state, producing immunoregulatory logically restrained in the face of trillions of commensal
cytokines such as IL-10; however, under inflammatory conditions they are a potent bacteria, but at the same time poised to defend the vast
source of inflammatory cytokines, including TNF, IL‑6 and IL‑1β. Tissue macrophages surface areas of the gastrointestinal tract from invading
exist as M1 (classically activated) and M2 (alternatively activated) subsets64. pathogens. Key cellular players of the mucosal immune
M1 macrophages are activated by IFNγ (a major product of the TH1 response), but also system include T cells, mononuclear phagocytes, innate
support TH1 activation or differentiation through IL‑12. IL‑4 is a major stimulus driving lymphoid cells and other cells of the innate immune
alternative activation of M2 macrophages. M2 macrophages are also a source of IL‑4, compartment (BOX 2; FIG. 1)63-72. Together, these cells com-
which supports TH2 cells65. Intestinal MPS cells are activated by bacterial products
municate with the epithelium, intestinal microbiota and
(such as lipopolysaccharide) that trigger Toll-like receptors (TLRs).
ENS. Crucially, immune function is both responsive to,
• Innate lymphoid cells (ILCs) are a heterogeneous population of newly described innate
and exerts influence upon, host stress responses.
lymphocytes that resemble the effector CD4+ lineages with respect to the expression
of lineage defining transcription factors, cytokine production profiles and functional The stress response is a rapidly mobilized, tightly
roles. Key subsets are ILC1 (express T‑bet and produce IFNγ), ILC2 (express GATA3 and coordinated, multisystem programme invoked in
produce IL‑5 and IL‑13) and ILC3 (express RORγt and produce IL‑22 and/or IL‑17A)66. response to adverse ‘stressor’ conditions, summarized
ILCs contribute to host defense against different mucosal pathogens, including in BOX 3 and reviewed elsewhere73. The stress response
helminths (ILC2) and bacterial infections (ILC1 and ILC3)66,67. IL‑22‑producing ILC3 regulates multiple aspects of immune function and, in
also serve to promote epithelial stem cell growth and recovery from injury, indicating return, several immune mediators are involved in trigger­
that these cells are crucial to long-term intestinal epithelial homeostasis68,69. ing the stress response and modulating its effects on dif-
• Immune-cell trafficking to the gut is dependent on the expression of selective homing ferent organ systems, including the gut. Neurons from
molecules, including the chemokine receptor CCR9 and the integrin α4β7 (REF. 70). the paraventricular nuclei (PVN) activ­ate the sympa­
The regulation of immune cell trafficking to the gut is now of major interest as thetic arm of the autonomic stress response, includ-
monoclonal antibodies blocking these pathways have emerged into clinical practice ing direct innervation of immune t­ issue in the gut,
to treat intestinal inflammation71,72.
including mesen­teric lymph nodes and gut-associated

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

Gut microbiota

Immunity Immunity Immunity to Epithelial


to bacteria to helminths bacteria and/or fungi resitution

Intestinal Histamine Eosinophils


epithelium TNF Neutrophils
Mast
cell Eosinophil
Macrophage granule
IL-4 proteins
IL-5
IFNγ IL-13 IL-5 IL-17 IL-22
TH1 ILC1 TH2 ILC2 TH17 ILC3 NCR+
ILC3
T-bet T-bet GATA3 GATA3 RORγt RORγt RORγt
IL-12

M2 macrophage

M1 macrophage

IFNγ IL-4

Monocyte
Figure 1 | Mucosal immune networks. The mucosal immune system has a crucial role in immunity to microorganisms
Nature
and in epithelial restitution. Distinct arms of host immunity can be mobilized to Reviews | Gastroenterology
defend against & Hepatology
specific microbial threats.
However, selective aspects of these immune response pathways can be inappropriately deployed in immune-mediated
diseases. T-Bet-expressing type 1 T helper (TH1) effector memory T-cells and type 1 innate lymphoid cells (ILCs) respond to
cytokine signals (including IL‑12) to trigger their effector responses, which includes production of IFNγ, which in turn
primes tissue mononuclear phagocytes, such as macrophages and monocytes, programming them for pro-inflammatory
activation. This pathway is especially important in host resistance to intracellular bacteria, including mycobacterial
infection. IL‑12‑producing M1 macrophages support the type 1 inflammatory response, and reciprocally IFNγ supports
M1 differentiation. The type 2 response is characterized by activation of GATA3‑expressing type 2 T helper (TH2) cells
and ILC2, which produce cytokines (such as IL‑4, IL‑5 and IL‑13) to support the activation, recruitment and survival of
eosinophils and mast cells. The type 2 response is implicated in host resistance to helminths. RORγt-expressing type 17
T helper (TH17) and ILC3 (especially natural cytotoxicity receptor (NCR)– ILC3) cells produce IL‑17 family cytokines,
which support the activation and recruitment of neutrophils and have an important role in host immunity to extracellular
bacteria and fungi. NCR+ ILC3 produce IL22, which supports the epithelial stem cell niche by promoting proliferation
of LGR5+ stem cells and also promote the production of antimicrobial peptides by epithelial cells. IL‑22‑producing ILC3
are implicated in host resistance to some mucosal pathogens.

lymphoid tissue (GALT)74. Sympathetic modulation of Moreover, adoptive transfer of the CD4+ compartment
immune t­ issue is focused around zones heavily popu­ to immunodeficient mice could reinstate colitis vulner-
lated by T cells, such as the cortical and subcortical ability in this stress model, confirming the key role of
areas in lymph nodes, with relative sparing of B‑cell- CD4+ T cells in driving s­ tress-induced gut dysfunction38.
rich ­germinal centres74. Noradrenergic fibres extensively In human IBD, acute stress leads to rapid increases in
arbor­ize in the interdomal regions of the lamina propria, rectal immune activation, including increased mucosal
which is especially extensive around T‑cell zones75. This production of TNF and the generation of reactive oxygen
direct line of communication permits rapid and focused species76. Stress-induced mucosal immune activation
modulation of immune function, and especially T cells could in part explain the increased intestinal perme­
through release of noradrenaline at this nerve-immune ability observed in individuals in whom public speak-
‘synapse’. These data reinforce important work in rodents ing triggers increased cortisol production77. To enable
that have shown a key role for CD4+ T cells in mediating immune cells to respond to nerve-derived signals, they
stress-induced gut dysfunction. Stress induction through express a variety of different receptors to neurotrans-
acoustic stimuli or restraint markedly increases colonic mitters, including dopamine78, serotonin (5‑HT)79,80,
permeability and reduces the threshold for reinduction of neuropeptide Y81, substance P82 and vasoactive intesti-
colitis by administration of intrarectal DNBS38. Crucially, nal peptide83, although work has mainly focused on the
this phenomenon could be recapitulated in CD8+ T‑cell- role of catecholamines in modulating immune function,
deficient mice, but not in CD4+ T‑cell-deficient mice. mostly through the β2 adrenergic receptor (β2AR)84–87.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
7
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
.
REVIEWS

Box 3 | The HPA axis and the stress response73 and resided in intimate contact with enteric nerves,
expressing an M2 transcriptional profile. By contrast,
• Activation of the stress response affects both brain and gut function and profoundly lamina propria macrophages expressed an M1 gene
influences dialogue between these organ systems expression footprint. M2 differentiation of muscularis
• After exposure to perceived threat, sensory inputs are directed and processed macrophages was dependent on β2AR, highlighting the
by limbic system structures in the forebrain, including the amygdala, hippocampus importance of nerve-mediated instruction of immune
and prefrontal cortex cell differentiation.
• The amygdala directly innervates the hypothalamus stimulating release of This process could be particularly relevant in FGIDs,
corticotropin-releasing hormone into the hypophyseal portal system, triggering for which data continues to amass regarding possible
release of adrenocorticotropic hormone from the anterior pituitary gland
­activation of type 2 immunity, including tissue eosino­
• Adrenocorticotropic hormone stimulates cortisol production by the zona fasciculata philia in FD and mast cells in IBS (discussed later).
of the adrenal cortex
Noradrenergic promotion of intestinal muscularis
• The amygdala also innervates the catecholaminergic locus coeruleus in the M2 macrophage differentiation or activation might
brainstem, which is the principle source of noradrenaline in the brain, serving
be ­capable of supporting type 2 immunity in the gut.
to augment arousal, focus and attention
At other mucosal surfaces, M2 macrophages directly
• Immediate sympathetic activation occurs after integration of stress signals
promote type 2 immunity through activation of TH2
at the paraventricular nuclei (PVN) of the hypothalamus, which can be considered
T cells, IL‑13‑producing innate lymphoid cell (ILC)2
the command centre of the autonomic stress response
and IL‑33‑dependent eosinophil recruitment 92,93.
• Neurons from the PVN project to sympathetic targets in the brain stem, spinal cord
Interestingly, duodenal eosinophilia observed in patients
and preganglionic noradrenergic nuclei to activate the sympathetic arm of the
autonomic stress response at target organs; direct and indirect (via synapses at the with FD is also characterized by infiltration of eosino-
nucleus tractus solitarius) projections exist to the locus coeruleus for induction of phils in the duo­denal submucosal plexus94. Additionally,
centrally acting noradrenaline release gastrointestinal pathogen exposure selectively drives
• Neurons from the PVN also directly innervate immune tissue in the gut, including further activation of muscularis-resident M2 macro­
mesenteric lymph nodes and gut-associated lymphoid tissue phages in mice, and it is tempting to speculate that
• The adrenal medulla is directly innervated by post-ganglionic sympathetic fibres, selective induction of type 2 immunity promoting M2
stimulating catecholamine release into the bloodstream; the adrenal medulla macrophages could potentially support mast cell (IBS) or
predominantly produces adrenaline and, to a lesser extent noradrenaline, eosinophil (FD) expansion and/or activation as observed
whereas the postsynaptic sympathetic fibres innervating the gut (and other organs) in post-infection FGID. M2 macrophage expansion
are noradrenergic and/or activation is observed at other mucosal surfaces
HPA, hypothalamic–pituitary–adrenal. in the context of TH2‑mediated disease95. Catecholamines
could also affect other aspects of immune function,
including humoral immunity, l­eukocyte ­development
Importantly, β2AR is only expressed by type 1 T helper and leukocyte trafficking 74.
(TH1) cells but not other T‑cell lineages84 and β2AR liga- ILCs also form important interactions with enteric
tion in TH1 cells suppresses cytokine production and glial cells. The outcome of this interaction has profound
activation markers88. Moreover, β2AR agonism impairs implications for epithelial homeostasis in the gut, the
in vitro and in vivo TH1 differentiation by suppressing the composition of microorganisms colonizing the gastro­
production of the critical crucial TH1‑skewing cytokine intestinal tract and host susceptibility to intestinal inflam-
IL‑12 by dendritic cells and monocytes87. As TH1 cells mation. Neurotrophic factors are proteins prod­uced by a
negatively regulate induction of other T‑cell effector sub- variety of tissues to support development, differenti­ation,
sets, β2AR‑ligation-induced suppression of IL‑12, which maintenance and survival of neurons. The extended
is mediated through inhibition of NF‑κB and tran- family of neurotrophic factor proteins include glial-cell
scription factor AP‑1 (REF. 89), indirectly favours type 2 derived neurotrophic factor (GDNF) and GDNF family
T helper (TH2)87 and type 17 T helper (TH17)89 differen- ligands (GFL), such as neurturin, artemin and persephin,
tiation. Interestingly, single nucleotide polymorphisms all of which are ligands for the tyrosine-protein kinase
at the ADRA1D locus encoding the adrenergic 1D receptor RET96. Surprisingly, using RET reporter mice,
receptor, which is also involved in sympathetic signal- intestinal ILC3s have been shown to also express RET97.
ling, are associated with severe IBS symptoms90, lending In the gut, enteric glial cells formed stellate projections
further support to the possibility that ­catecholaminergic to interact with ILC3 in cryptopatches. These glial cells
­signalling is important in gut symptoms. were a major source of GFL, which was a power ­trigger
Data has also cast new light on the potential role of the to induce IL‑22 by ILC3. Selective deletion of RET in
stress response and catecholamine signalling between ILC3 resulted in severely diminished IL‑22 produc-
the ENS and the innate immune system that would also tion by ILC3, reduced production of antimicrobial
favour type 2 immunity in the gut. Using 3D imaging of defences, dysbiosis, increased bacterial translocation
intact intestinal tissue and in vivo imaging of mice with across the undefended gut wall and, ultimately, height-
fluorescent tags marking either macrophages or enteric-­ ened susceptibility to gastro­intestinal infections and
associated neurons, Gabanyi et al.91 demonstrated a intestinal inflammation97. Crucially, enteric glial cells
gradient in the functional specialization of intestinal produce GFL in response to Toll-like receptor (TLR)
macrophage networks across the gut wall and their agonists or the alarmin cytokines IL‑1β and IL‑33 in a
interaction with enteric nerves. Macrophages in intes- MYD88‑dependent manner 97. These data indicate that
tinal muscle layers exhibited distinctive morphology enteric nerves can respond to presence of microorganisms

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

through TLR sensors, or early cytokine stimuli to trigger favour of low-grade inflammation contributing to the
ILC3 activation through production of GFL acting on the aetiology and symptomatology of FGIDs grows ever
RET receptor. Key interactions between the gut, immune more persuasive. Notably, there is a high prevalence of
system and brain are depicted in FIG. 2. functional gastrointestinal symptoms in patients with
recognized inflammatory diseases of the gut, including
Immune activation in FGID. Although FGIDs are con- IBD, infectious gastroenteritis, coeliac disease and micro-
ventionally defined by the absence of clear-cut patho­ scopic colitis even after sustained resolution of overt
logical changes, such as overt macroscopic inflammation, inflammation58,98–101. Moreover, patients with FGIDs are
a shift in momentum has emerged to challenge this view- at increased risk of developing other immune-mediated
point. Numerous studies, although not all, have identified diseases102, consistent with a possible immune diathesis
subtle immune changes in the gut of patients with FGIDs in FGID. Numerous studies have described increased
(TABLE 1). Although ongoing debate remains regarding pro-inflammatory cytokines (for example, IL‑6, TNF and
the relevance of these findings as primary pathological IL‑1β) in the serum or plasma of patients with FGID,
events, or bystander epiphenomena, the evidence in or increased production of these cytokines following

Stress response

Gut microbiota

AMP
Intestinal
epithelial
cell IL-22R IL-22 Mast Histamine
IL-4 cell
ILC3
Lamina RET
propria
GFL TH2 cell
IL-5
Enteric β2AR
neuron Eosinophil Eosinophil
NA
granule proteins
Circular
muscle

Myenteric ENS
plexus

M2 macrophage

Longitudinal
muscle

Systemic circulation
Cytokine-producing cells
(IL-1β, TNF, IL-6)
Figure 2 | Key brain–immune–gut interactions. There is complex crosstalk between the immune system, the brain
and the gut. Noradrenergic neurons in the gut release noradrenaline (NA),Nature which Reviews | Gastroenterology
ligates the & Hepatology
β2 adrenergic receptor (β2AR)
on macrophages in the myenteric plexus (supporting differentiation towards the M2 phenotype) and T cells, which limits
T helper (TH)1 differentiation (indirectly favouring TH2 and TH17 differentiation). Mast cells and eosinophils are found
degranulating next to enteric neurons, providing a mechanism for sensory excitation (which can be perceived by the
enteric nervous system (ENS) and central nervous system). Gut immune cells (e.g. γδ T cells) differentiating in the gut can
traffic to the brain under some circumstances (e.g. after brain injury). Blood-borne cytokines generated in the gut
can also signal in the brain. Enteric glial cells also produce glial-cell-derived neurotrophic factor family ligands (GFL),
which stimulate IL‑22 production by innate lymphoid cell (ILC)3 (acting on the specific receptor RET). IL‑22 acts on
an epithelial‑restricted receptor (IL‑22R) to stimulate epithelial proliferation, antimicrobial peptide production.
IL‑4 and IL‑5 (potentially generated locally by TH2 T cells) support activation of mast cells and eosinophils, respectively.
AMP, antimicrobial peptides.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
7
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
.
REVIEWS

short-term culture of peripheral blood leukocytes iso- histamine H1 receptor (HRH1), as the effect could
lated from patients with FGID, often at concentrations be blocked by HRH1 antagonists and were blunted in
correlating with disease features or psychological mor- ­neurons harvested from Hrh1−/− mice119. Mast cells are
bidity 103–108. Some studies have detected mucosal abnor- also implicated in loss of intestinal barrier function in
malities in patients with FGID. Unsupervised profiling of stress models. A marked expansion of colonic mast cells
differentially expressed genes in colonic biopsy samples and increased intestinal permeability is observed in rats
from patients with IBS and healthy individuals shows following stress induction by water deprivation; how-
that the majority of differentially expressed genes localize ever, the permeability defect can be rescued when stress
to the mucosal immune system109,110. is induced in m­ ast-cell-deficient animals (Ws/Ws rats)120.
In IBS, arguably the most compellingly evidence A similar picture is emerging in FD, although rather
for a role of disordered immunity comes from studies than primarily mast cells, a disordered homeostasis
examining mucosal mast cells. Increased numbers of of eosinophils in the proximal small bowel also seems
mucosal mast cells have been observed in patients with involved (FIG. 3b,c). In a landmark study of non-health-
IBS62,111–116 (FIG. 3a), often correlating with key symptoms care seeking individuals from Sweden who completed
such as abdominal pain111,115 or bloating 116, and might validated gastrointestinal symptom questionnaires and
be seen degranulating in close proximity to enteric underwent upper gastrointestinal endoscopy, eosino-
nerves111. Production of mast cell products, including phils were found to markedly increased in the proximal
tryptase, histamine and prostaglandins, is increased in duodenum in 40% of individuals reporting FD symp-
gut tissue from patients with IBS111,117, and jejunal fluid toms121. Subsequent work has corroborated these find-
harvested from patients with diarrhoea-predominant ings and consistently links early satiety, a hallmark FD
IBS have increased levels of tryptase112. In vivo admin- symptom, with expansion of duodenal eosinophils122–125.
istration of supernatants derived from explant cultures In patients with FD, accumulating duodenal eosinophils
from patients with IBS to the artery supplying a loop of are activated, degranulating in close proximity to enteric
jejunum in experimental rats induced marked excitability nerve fibres.These observations have been extended,
of visceral sensory nerve fibres and triggered excessive showing that eosinophils and mast cells are massively
calcium flux in dorsal root ganglia, effects which were expanded in the duodenal submucosal plexus of patients
not observed following administration of explant culture with FD94. In this elegant study the researchers were able
supernatants from control individuals117. Submucosal to quantify neuronal activity by measuring calcium flux
nerves harvested from the distal colon of Guinea pigs also in submucosal nerve fibres. Although the number of
exhibit exaggerated excitability when exposed to culture nerves and ganglia present in the submucosa of patients
supernatants derived from explants from patients with with FD was comparable to healthy individuals, there was
IBS118. Consistent with mast cell degranulation driving ­striking impairment of nerve excitability in FD. Moreover,
symptoms in IBS, histamine potentiates triggering of blunting of submucosal nerve fibre excitability correlated
colonic submucosal neurons with the TRPV1 agonist with eosinophil and mast cell infiltration, highlighting
capsaicin119. Histamine potentiated colonic nocicep- the potential importance of interactions between enteric
tive afferent nerve fibres were mediated through the nerves and immune cells in the pathogenesis of FD.

Table 1 | Evidence for immune dysregulation in FGID


Focus Evidence
Epidemiology • High prevalence of ‘functional’ symptoms in patients with overt gastrointestinal inflammation,
including IBD (even in remission) and coeliac disease58,98–101
• Increased prevalence of other autoimmune diseases in patients with FGID102
Mast cells • Mast cells and their degranulation products (e.g. histamine, tryptase) are increased in colon or small
bowel of patients with IBS62,111–117
• Tissue mast cell numbers correlate with IBS symptoms111,115,116
• Antihistamines and mast cell stabilizers are effective in IBS119,204,205
Eosinophils Increased numbers of eosinophils in the duodenal mucosa and submucosal plexus, which correlate
with presence of functional dyspepsia symptoms94,121–125
Lymphocytes • Increased numbers of intraepithelial lymphocytes in IBS62,112,115,129
• Increased numbers of gut-homing lymphocytes in peripheral blood of patients with FGID,
which correlate with symptoms107,128
• Circulating lymphocyte numbers rapidly decrease in the postprandial phase in IBS130
Nerve/immune • Mast cells can be observed degranulating in close proximity to enteric nerves111
interactions • Histamine potentiates firing of colonic pain afferents119
Cytokines • Increased levels of cytokines in serum or plasma, or following short-term culture of peripheral blood
leukocytes from patients with FGID103–108
• Dysregulated T‑cell cytokine responses (shift to type 2 T-helper responses) in FGID127
Genomics • Immune genes are differentially expressed in gut biopsy samples from patients with IBS109,110
• Polymorphisms at loci encoding immune (IBD-associated) loci in IBS (e.g. TNFSF15) 216,217
FGID, functional gastrointestinal disorder.

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

consistent with the possibility of increased gut homing


a b
of pathological T‑cell lineages in IBS. Some studies have
also identified increased numbers of gut T cells in IBS,
particularly in the intra­epithelial compart­ment62,112,115,129.
Consistent with gut lymphocyte trafficking being
influenced by food intake, in patients with IBS a rapid
reduction in the number of circulating lymphocytes in
the post-­prandial phase is seen130. The emergence of
ILCs as mediators of intestinal inflammation66 raises
the ­intriguing possibil­ity that these cells might also be
involved in FGIDs, and especially the ILC2 subset that
c produces type 2 cytokines capable of supporting and
activating mast cells and eosinophils. Notably, IL‑1β,
which is excessively produced by peripheral blood
mono­nuclear cells from patients with FD107, activates
ILC2 to produce IL‑5 (REF. 131). The potential role of
ILCs in FGIDs is currently being addressed.
Although local mucosal immune activation might
plausibly be expected to affect gastrointestinal physio­
logy (and ultimately symptoms) through interaction with
enteric nerves and gut smooth muscle, cytokines prod­
uced by immune cells might also act centrally to affect gut
function. For example, IL‑1β and TNF-mediated inhib­
ition of gastric emptying is achieved at much lower doses
when these cytokines were administered intra­cisternally
(compared with intravenously), is dependent on an intact
vagus nerve, and in the case of IL‑1β, is reversible by
intracisternal IL‑1β antagonists132,133.
Figure 3 | Low-grade inflammation inNature | Increased
FGID. aReviews numbers of tryptase-positive
| Gastroenterology & Hepatology
mast cells (brown staining) in the colon of patients with IBS. b | Expansion of duodenal Immune regulation of brain function
eosinophils in the duodenal mucosa of patient with functional dyspepsia. c | Eosinophil Extrinsic and intrinsic routes. Ostensibly, the immune
degranulation in duodenal mucosa of a patient with functional dyspepsia (carbol system has at least three avenues of communication
chromotrope stain, arrow) adjacent to nerves (S100 immunostaining, arrowhead). to conduct dialogue with the brain. Firstly, cytokines
Inset: intact eosinophil. Magnification for parts a and b ×20, for part c ×40 and inset ×100. (or hormones) generated by immune cells or other line­
FGID, functional gastrointestinal disorder. ages in the gut (or elsewhere in the periphery) could be
transported in the blood to act on the brain (humoral
The underlying factors responsible for initiating and route). Other potentially relevant humoral factors
sustaining duodenal eosinophil and mast cell accumu­ include bacterial cell wall products, such as lipopoly-
lation in patients with FD have yet to be resolved, saccharide, as these ligands can trigger TLRs expressed
although it has been hypothesized that dysregulated by microglial cells in the CNS (discussed later). Second,
CD4+ TH2 responses could be responsible, as these immune cells in the gut could directly communicate
cells are potent producers of key cytokines involved in with afferent sensory fibres and/or enteric nerves, often
supporting the recruitment, survival and activation of through production of cytokines or other immune
eosinophils and mast cells, such as IL‑4, IL‑5 and IL‑13 mediators (such as hista­mine) to trigger signals that
(REF. 126). Furthermore, there is experimental support are ultimately relayed to the brain via neurons. Third,
for this hypothesis, as polyclonal stimulation of T cells circulating immune cells have the potential to ­traffic
isolated from patients with FD results in markedly to the brain, where they can liberate cytokines and
increased prod­uction of IL‑5 and IL‑13, with a recipro­ other inflammatory mediators in close proximity to
cal reduction in the production of the TH1 cytokine relevant brain structures. T‑cell trafficking from the
IFNγ127. Consistent with a functionally important role gut to the brain has been elegantly demonstrated using
for T cells in FGIDs, T cells expressing gut-homing KikGR33 mice that constitutively express Kikume green-
integrins and chemokine receptors, including α4β7 red (KikGR) fluorescent protein that converts from a
and/or CCR9, are markedly increased in patients green to red fluorescent tag after exposure to ultraviolet
with IBS128 or FD107. In the case of FD, the number of light. T cells in the intestine were photo­converted into
α4β7+CCR9+CD4+ T cells is p ­ ositively corre­lated with red-­fluorescent-tagged cells by exposing the small intes-
FD symptoms (such as abdominal pain, ­nausea and tine to UV light at laparo­tomy. However, red-­fluorescent-
vomit­ing) and with reduced gastric empty­ing, measured tagged T cells that could only have been generated in
by scinto­graphy 107. Similar to the situ­ation in acute ulcer­ the intestine were also recovered from the meninges
ative colitis, MAdCAM1, the ligand for the gut-­homing and cervical lymph nodes after ischaemic injury to
integrin α4β7, is markedly upregulated in the colon the brain134. These data confirm that, under defined
of patients with IBS relative to healthy individ­uals128, inflammatory settings, T cells can traffic from the gut to

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
7
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
.
REVIEWS

the brain. Intriguingly, T cells destined to traffic to the and/or function and inappropriate activation of brain
gut or the brain share expression of the tissue-homing myeloid cells, including microglia, plays a central part in
­integrin α4, although they have distinct β chains70. neuroinflamamtion and are strongly implicated in other
Overall, the most evidence has been amassed regard- brain diseases, including multiple sclerosis (MS) and
ing how cytokine networks modulate brain function. Alzheimer disease (AD). In MS, the relative contribution
Cytokines are a family of >100 small protein molecules of microglia and other cells of the MPS remains contro-
that can access the brain through regions of the blood– versial. On the one hand they are key reparative agents in
brain barrier (BBB) that are prone to leakiness. Access the brain involved in restitution following injury (includ-
occurs through active uptake, modulation of the vascular ing experimental demeyelin­ation), versus their potent
endothelium of vessels supplying the brain and by acting pro-inflammatory roles as producers of harmful effector
on peripheral vagal afferents in the gut, which in turn cytokines (for example, TNF), and cytokines involved in
relay signals to pertinent nuclei in the brain, including the differentiation and maintenance of pathogenic TH17
the nucleus tractus solitarius (NTS) and hypothalamus135. and TH1 cells (for example, IL‑1, IL‑6, IL‑23 and IL‑12)143.
Cytokines might also act centrally to potentiate sensory Microglia and other MPS cells also contribute to clearance
afferent inputs from the gut. Indeed, in animal models, of extracellular amyloid‑β deposits, and disruption in the
TNF activates the neurons at the NTS that are respon- balance between amyloid‑β accumulation and micro-
sive to gastric distension136. Although many different glial or MPS-mediated removal is probably a key patho­
cytokines have been shown to modulate brain activity, logical event in the development of AD. CD14 (the TLR4
IL‑6, TNF and IL‑1β might be regarded as the ‘unholy co‑­receptor) expressed by microglial cells directly inter-
trinity’ of cytokines that affect brain function at multi- acts with amyloid‑β and facilitates its clearance through
ple levels. These cytokines are also known to activate the phagocytosis144. Why this process is impaired in AD is
sympathetic nervous system and HPA axis, which in turn currently under scrutiny, although impaired expression
have direct effects on the brain74. of TLRs by microglia in AD and its potential link to defec-
In addition to these ‘extrinsic’ routes of immune regu­ tive phagocytosis of amyloid‑β is currently being actively
lation of brain function, it is also important to realise pursued. How the role of microglia and other immune
the importance of brain ‘intrinsic’ immune regulation. cells resident in the CNS behave and contribute to brain
Microglial cells are the key immune cells of the CNS. dysfunction in systemic diseases, particularly to gut dis-
These cells are distributed throughout the brain and spinal eases including FGIDs, is currently unknown. However,
cord, but are especially abundant in the grey matter, where it might be expected that TLR engagement could result
they extensively branch and communicate with neurons from impaired gut barrier function and bacterial trans-
and other cells of the CNS. Microglia differentiate from location in numerous gut diseases. Increased intestinal
the same immediate haematopoietic precursor as the permeability is observed in IBD145 and in FGIDs146, which
closely related monocyte–macrophage lineage. However, is associated with increased concentrations of bacterial
unlike intestinal macrophages, which are continually products, including the TLR agonists LPS (TLR4) and
replenished by circulating monocytes, the microglial flagellin (TLR5) in the serum of these patients147,148.
compartment is mostly maintained by self-renewal or
in situ proliferation, although in overwhelming infection Cytokines modulate mood. The role of cytokines in mood
or injury, microglial replenishment and/or expansion can disorders has been intensively studied, and even the
be supported by circulating monocytes137. As might be possibil­ity that these agents might serve as primary caus-
expected, microglia share functional character­istics with ative agents in depression has been debated149. Some have
other cells of the mononuclear phagocyte system (MPS). shown dysregulated cytokine levels in biofluids, including
For example, they are phagocytic, express TLRs and serum, plasma and cerebrospinal fluid of patients with
MHC class II molecules, and produce pro-­inflammatory severe idiopathic depressive symptoms135. Other immune
cytokines, particu­larly in the activated state138,139. Microglia changes include shifts in the numbers of monocytes,
respond to TLR engagement by initiating potent cytokine neutro­phils and lymphocytes, as well as T‑cell activation
responses. Ligation of TLR3 triggers microglia to produce status and serum complement levels150,151. Patients treated
TNF, IL‑6 and IL‑12 (REF. 139), highlighting the critical with cytokines for other disease states are also prone to
role of brain immune cells in sensing threat and initiating developing depression. For instance, IFNα administered
inflammatory ­cascades. Some of these responses might be to treat hepatitis C frequently induces depressive symp-
important for shaping neuronal repair following injury. toms152 and patients with increased baseline serum TNF
Indeed, consistent with a neuroprotective role of TNF, levels are especially susceptible153. These observations are
genetic deletion of this cytokine (Tnfa−/− mice) results in supported by mouse studies in which administration of
impaired regener­ation of oligo­dendrocytes after toxin-­ recombinant cytokines, or the immune activator LPS,
induced demyelin­ation140. In addition to the reparative induces sickness behaviour in experimental animals with
roles of microglial cells, innate immune sensing of patho­ features of depression154. For example, in rodents, intra­
gens through TLRs has a central role in host resistance cerebral injection of recombinant IL‑1β or TNF induces
to brain infections, including pneumococcal meningitis pyrexia, sleepiness and hyperalgesia in a dose-­dependent
(TLR2 and TLR4)141, herpes simplex virus (HSV) and manner 155,156. IL‑6, TNF and IL‑1β in particular are also
encephalitis (TLR3)142. Indeed, otherwise healthy c­ hildren potent activators of the stress response, activating the
carrying dominant-­negative TLR3 alleles are suscep­ HPA axis at multiple levels, including activ­ation of neu-
tible to HSV infection142. Dysregulated TLR expression rons containing corticotropin-releasing hormone in the

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

PVN of the hypothalamus, and even direct activation possesses anti-inflammatory properties in vitro and
of the adrenal cortex 157,158. Consistent with the prevail- in vivo, potentially explaining why its absence has been
ing view that depression is closely linked to disordered linked to worse outcomes in patients with Crohn's dis-
neurotransmitter homeostasis, cytokines (again, mostly ease167. Reduced intestinal microbiota diversity is also
IL‑1β, IL‑6 and TNF) have been shown to regulate a feature of IBS169–171 and has been linked to expansion
multiple different neurotransmitters, including central of gut lymphocytes and increased ­anxiety 170. Other
noradrenergic (predominantly IL‑1β), dopaminergic and data support a shift in the relative proportion of major
serotonergic systems149. bacterial phyla in some patients with IBS, character­
ized by a relative expansion of Firmicutes and contrac-
Cytokines disrupt blood–brain barrier function. tion of Bacteroidetes172. Interestingly, F. prausnitzii, the
Maintenance of the microvascular endothelial barrier bacterium that seems to provide anti-­inflammatory
has an important role in regulating brain function, and functions in IBD, is also depleted in patients with
cytokines play an important part in regulating this process. diarrhoea-predominant IBS169.
TNF and IL‑6 increase the permeability of brain micro­
vascular endothelial cells, which culminates in disrup- Gut microbiota regulate key aspects of host immunity.
tion of the BBB159,160. Mechanistically, this process occurs The composition of intestinal microbiota has an impor-
through induction of reactive oxygen species and NF‑κB, tant role in shaping host immunity, including influences
which repress tight junction proteins, such as occludin, over cytokine expression, both in the gut and in sys-
claudin‑5 and zonula occludens‑1, at the transcriptional temic tissues. Germ-free mice have numerous immune
and protein level in a dose-dependent ­manner 159,161,162. abnormalities, including grossly enlarged caeca, failure
High-dose TNF might even directly ­trigger brain micro- of second­ary lymphoid development (for example, lym-
vascular endothelial apoptosis in vitro160. Importantly, the phoid follicles), impaired antibody responses, diminished
BBB itself is an immune-active compartment and astro- numbers of T cells and B cells and defective production of
cytes, which have a key role in the formation and mainten­ cytokines (such as TNF, IL‑6 and IL‑1β)173,174. Moreover,
ance of the BBB, express TLRs, enabling them to sense selective constituents of the gut microbiota shape speci­
and respond to microbial exposure. Engagement of TLR3 fic aspects of adaptive and innate immunity, including
­triggers astrocytes to produce cytokines, such as IL‑6, the differentiation of particular effector T‑cell lineages.
IFNβ but also IL‑10 (REFS 139,163). For example, unless mice are colonized with bacteria that
can adhere to intestinal epithelial cells, such as segmented
The microbiota–gut–brain axis fila­mentous bacteria Citrobacter rodentium or E. coli
Molecular characterization of the microbial communi­ 0157, they fail to mount effective TH17 responses175,176.
ties occupying the healthy human gastrointestinal Other bacteria, including Listeria, are permissive for host
tract demonstrates that >90% of the species occupy- TNF, IL‑1β and TH1 responses176–179. Conversely, some
ing this ecological niche belong to two main phyla, the bacteria promote immunosuppressive cytokines, such as
Firmicutes and Bacteroidetes, with minor contributions IL‑10 by regulatory T cells, which can be dependent on
from Actinobacteria and Proteobacteria164. However, bacterial prod­uction of short-chain fatty acids (SCFAs)
the composition of intestinal bacterial communities is by ­clusters of Clostridia180,181, or polysaccharide A derived
disrupted in IBD and FGIDs, and some aspects of the from Bacteroides fragilis 182. Together, these data show
gut microbiota crucially regulate selective immune that the precise composition of the intestinal microbiota
response networks. can qualitatively and quantitatively alter host immune
responses, with clear implications for how this step might
The gut microbiota community structure is disrupted influence the concentration and profile of cytokines
in IBD and FGIDs. Interactions between the microbial present in any given individual, which in turn has the
communities that colonize the human gastrointestinal capacity to differentially affect brain function. Microbial
tract and their host have far-reaching consequences for influences over T‑cell differentiation, including lineage
host health, including governing susceptibility to disease. fate decisions, are illustrated in FIG. 4.
In a number of disease states, including gastro­intestinal
disease, there are shifts in the community structures of The intestinal microbiota modulates brain function.
the intestinal microbiota and growing evidence points to Mounting evidence highlights the importance of the
the likelihood that these changes are not epi­phenomena intestinal microbiota in modulating multiple features
occurring secondary to the underlying disease pro- of brain function, including the HPA-axis-regulated
cess, but instead have integral roles in disease patho- stress response, emotional responses, memory, motor
genesis and/or clinical course. The dys­biosis present in behaviour and aspects of social interaction. One of the
patients with IBD is character­ized by loss of bacterial key areas of study to date concerns the reciprocal role
diversity, contraction of some phyla, such Bacetroidetes of the intestinal microbiota and the stress response.
and Firmicutes, and expansion of others (such as Stress alters the community structure of the gut micro-
Proteobacteria)165. Some s­ pecies, such as adherent-­ biota183, but experimentation with germ-free rodents has
invasive Escherichia coli have been pathologically again provided clear evidence that the intestinal micro­
implicated166. Other species, such as Faecalibacterium biota directly affects stress responses and thresholds for
prausnitzii and Roseburia ­hominis are selectively depleted anxiety. Pioneering work by Sudo et al.184 convincingly
in patients with IBD167,168. Interestingly, F. prausnitzii demonstrated that the intestinal microbiota substantially

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
7
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
.
REVIEWS

Gut microbiota In addition to reduced anxiety, other key behav-


ioural and cognitive changes observed in germ-free
SFB SFB Bacteroides fragilis animals include increased motor activity, impaired
Listeria Citrobacter rodentium Clostridial clusters memory and defective social functioning, including
Escherichia coli 0157
reduced time spent in social investigation and increased
engagement of repetitive grooming activity during
social interaction187,192,193. Data has demonstrated how
the intestinal microbiota influences brain injury, and
moreover, has identified the mucosal immune system
Intestinal as the key driver of this phenomenon. By reducing
TH1 TH17 Treg epithelium
the complexity and diversity of the gut microbiota
with antibiotics, treatment for brain infarct volume is
IFNγ IL-17A
markedly reduced in mice following transient middle
TNF
cerebral artery occlusion134. Crucially, protection from
ischaemic injury was conferred by defective differ-
M1 macrophage Neutrophils entiation of microbially induced, pro-inflammatory
intestinal γδ T cells, and a failure of these cells to then
Figure 4 | The gut microbiota directlyNature
influences T‑cell
Reviews differentiation. Specific
| Gastroenterology & Hepatology traffic to the brain after injury. In mice colonized with
bacterial constituents of the gut microbiota are required for lineage differentiation of conventional, diverse bacterial communities, robust
different T‑cell subsets. Bacteria, including segmented filamentous bacteria (SFB), differentiation of IL‑17‑producing γδ T cells in the gut
Citrobacter rodentium and Escherichia coli 0157 (which are all epithelial adherent was found. These cells were then observed to traffic to
bacteria) are permissive for T helper (TH)17 differentiation. SFB and Listeria are also
the brain following ischaemic injury, where they propa­
implicated in the differentiation of TH1 cells. Regulatory T‑cell (Treg) numbers are boosted
by the presence of selective clostridial clusters and Bacteroides fragilis. gated a pro-inflammatory programme that exacer­
bated tissue damage. However, in mice with reduced
bacterial diversity induced by antibiotic treatment,
affected host responses to stressful stimuli. A twofold impaired differentiation of IL‑17‑producing‑γδ T cells
increase in the magnitude of the HPA stress response, in the gut was found, with fewer of these cells migrat-
as measured by increases in plasma adrenocorticotropic ing to the meninges or cervical lymph nodes, thereby
hormone and corticosterone, was detected in germ-free limiting post-ischaemic inflammatory tissue damage.
mice following restraint-induced stress, in comparison Intriguingly, work indicates that ischaemic or traumatic
with conventionally colonized mice. Reconstitution brain injury itself can drive changes in the intestinal
of germ-free animals with probiotic bacteria, such as microbiota in mice 194. These changes were mostly
Bifidobacterium infantis suppressed the stress response, attributed to activation of injury triggered noradrener­
whereas monoassociation with enteropathogenic E. coli gic stress responses. Noradrenaline was increased in
(EPEC) further amplified the stress response, demon- the caecum following stroke and, moreover, the shifts
strating that it is not just the presence or absence of intes- in the composition of the microbial communities
tinal bacteria that influence the stress response, but also observed following stroke could be recapitulated by
qualitative compositional changes with individual ­species pharmacologically increasing noradrenaline release.
having profoundly different effects. Unlike B. infantis, This work corroborates other data showing how
reconstitution with EPEC triggered a notable increase the stress response markedly affects composition of the
in plasma IL‑6 and IL‑1β levels. Crucially, reconstitution gut microbiota.
with a mutant strain of EPEC that cannot bind to the
intestinal epithelium failed to trigger the same magni- Gut microbiota direct enteric neurogenesis. The intes-
tude of stress response and was unable to induce robust tinal microbiota serve to fine-tune the development of
IL‑6 and IL‑1β responses. Other work has corroborated a mature, functional ENS. The majority of the inter-
these findings and also investigated behavioural conse- connecting neural circuits of the outer myenteric plexus
quences associated with the germ-free state. Curiously, and the inner submucosal plexus are formed during
these experiments tend to show reduced anxiety in embryogenesis under the direction of specific transcrip-
germ-free animals185–187. Subsequent work has shown tion factors (such as SOX10, FOXD3 and PHOX2B),
that prolonged or short-term stressful stimuli disrupt crucially coupled in a coordinated developmental pro-
the community structure of bacteria present in both the gramme to the simultaneous formation of the other
faecal stream and colonic mucosa183,184,188–190, and specific key structures of the gut wall, including the epithelium,
changes induced include contraction or loss of beneficial muscle layers and lymphoid structures195. Integration
probiotic-like lactobacilli188–190 and expansion of particu- of these units into functional circuits continues post-
lar genera, including Coprococcus and Pseudobutyrivibrio natally and signals from intestinal microorganisms,
— coloniza­tion of which was associated with more pro- which rapidly colonize the gastrointestinal tract in the
nounced production of proinflammatory cytokines, such immediate postpartum period, help to shape the func-
as IL‑6 (REF. 183). Stressful stimuli might also facilitate tional maturation of this system. Germ-free mice have
colonization with overtly pathogenic bacteria, such as reduced density and number of nerves in the myentric
C. rodentium, which can induce a s­ ubstantial local plexus and diminished spontaneous smooth muscle
inflammatory response and overt colitis191. ­contractility in the gut wall196.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

The gut microbiota also regulate changes in the CNS. Therapeutic implications
As already discussed (summarized in BOX 1), the ENS The intimate, yet complex, interactions between the gut,
and CNS are highly integrated and major changes in brain, immune system and intestinal microbiota have
the growth and development of both of these systems far-reaching implications for host health. Looking for-
occur at a time that coincides with de novo population ward, the therapeutic horizon in FGID and IBD is likely to
of neo­natal gut with bacteria. Experiments in germ-free see interventions targeting these systems as primary treat-
animals have again highlighted structural and func- ments. These therapeutic paradigms and their p ­ otential
tional changes in the brain that are specifically linked to inter-relationships are represented in FIG. 5.
bacterial coloniza­tion of the gastrointestinal tract. Gut-
microbiota-regulated CNS structural changes are focused Psychological interventions. Psychological interventions,
in the limbic system, at centres associated with m­ emory, including psychotherapy, hypnosis and anti­depressant
behaviour and emotional response. For ­e xample, therapy, are effective in treating IBS197, although respon­
in germ-free mice, there is volumetric expansion of the ses are variable, in keeping with the possibility that some
amygdala and hippocampus, and individual neurons patients’ psychological distress precedes IBS (brain-gut
in these regions show striking morphological changes, directed) and in others it follows on from IBS (gut-
including dendritic hypertrophy of amygdala neurons198. brain directed). Cognitive behavioural therapy (CBT)
Indeed, early seminal work showed reduced expression shows promise as a psychological intervention in IBS199
of key neutrophins, such as brain-derived neurotrophic and large randomized controlled trials (ACTIB and
factor, in the hippocampus of germ-free animals184. IBSOS) evaluating CBT in comparison with conventional
care or placebo are underway and might yet inform fur-
ther novel evidence-based practice200,201. Active screening
Stress response to enable early identification of psychological disturbance
• Psychotherapy in FGIDs will probably be key to considering which
• CBT
psycho­therapeutic interventions are most likely to be
• Antidepressants
• Anxiolytics effective. Other strategies targeting stress alleviation are
also being considered, including self-­directed therapies
and mindfulness-based stress ­reduction, although robust
efficacy data are currently lacking.

Immune interventions. As inflammation could have a


role in driving gastrointestinal and psychological symp-
Disordered motility Immune activation
Visceral hypersensitivity • Anti-cytokine mAbs toms in FGIDs, it is tempting to speculate that targeted
• Antispasmodics • Antihistamines immunotherapies might be effective new treatments in
• Antidepressants • Topical steroids this arena. Notably, oral prednisolone had no effect on
• Autonomic blockade • Mast cell stabilizers PI-IBS symptoms (although it did reduce mucosal T‑cell
• Other
immunosuppressants
numbers)202. The efficacy of prolonged courses of high-
dose topical steroids remains to be seen. Likewise, it has
yet to be established whether selectively targeting individ-
ual cytokines with monoclonal antibodies will uncouple
the brain–gut axis and simultaneously treat inflam­
mation and inflammation-associated brain dysfunction.
Dysregulated
intestinal microbiota Biological agents targeting TNF, IL‑1β and IL‑6 among
• Antibiotics others, are already used in the clinic for a broad range of
• Probiotics inflammatory diseases63,203. Ongoing clinical trials inves-
• Prebiotics tigating blockade of individual mediators might provide
• FMT
• Dietary modification
insights into how they drive psychological aspects of
disease independently of their effects in resolving tissue
inflammation, and furthermore, might highlight the
effectiveness of these strategies at treating psychological
manifestations. Given the potential importance of mast
cells (IBS) and eosinophils (FD) in FGIDs, alternative
immune interventions targeting these cells might be
Figure 5 | Therapeutic paradigms for interfering with the brain–gut axis. expected to yield fruitful results. Historical studies have
Interventions directed at either the brainNature
or gutReviews
aspect of| Gastroenterology
the axis might be anticipated
& Hepatology shown possible efficacy of mast cell stabilizers in patients
to alleviate clinical features in both organ systems. For example, in patients in whom with IBS204,205, and a proof‑of‑principle pilot study in IBS
gut symptoms are primarily driving brain symptoms, modulation of the microbiota
antagonizing the histamine receptor HR1H has shown
(e.g. faecal microbiota transplantation (FMT) or probiotic therapy) would directly alter
host immune responses, alleviate local immune activation and simultaneously reduce promising results, including reduced abdominal pain
immune-driven brain dysfunction. Likewise, targeting the stress response and reduced visceral hypersensitivity in comparison with
(e.g. psychotherapy or antidepressants) would alleviate brain symptoms, but would placebo-treated patients119. In the future, antagonists that
also limit autonomic activation of host immunity to suppress aberrant immune-driven additionally target other histamine receptors respons­
gut symptoms. CBT, cognitive behavioural therapy; mAb, monoclonal antibody. ible for mediating central effects might confer additional

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
7
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
.
REVIEWS

benefits. For instance, H3 receptor antagonists show immune responses in IBD and FGIDs. From a preci-
promise as novel anxiolytics in preclinical models206 and sion medicine perspective, it is conceptually attractive
reagents targeting both H1 and H3 receptors might be to consider that individual patients will ultimately be
expected to simultaneously modulate immune activation stratified according to clinical phenotypes (pattern of
and stress responses in patients with IBS. Trials evaluat- functional symptoms and magnitude or profile of psycho­
ing more selective anti-allergy drugs, including mono- logical symptoms), together with biomarkers reflecting
clonal antibodies targeting IL‑5 or IL‑4–IL‑13, might be patterns of mucosal and systemic immune activ­ation,
expected to yield more impressive results, especially if as well as in‑depth profiling of intestinal bacterial com-
eosinophils are genuinely functionally important in FD. munities. Patients could then be stratified to specific
treatments based on implicated pathways. For instance,
Manipulation of the microbiota. Manipulation of the patients with prominent IBS symptoms and major anx-
microbiota through administration of probiotics, pre­ iety symptoms that are linked to selective enrichment
biotics, antibiotics (including highly selective agents of IL‑6 in serum and/or in mucosal responses might be
and nonabsorbable varieties), dietary modifications and expected to benefit from blockade of the IL‑6 pathway.
faecal microbiota transplantation (FMT) are promising Similarly, patients with reduced numbers of regu­latory
approaches in intestinal diseases, including FGIDs207–210. T cells, diminished IL‑10 responses and depletion of
Early work showed that monoassociation of germ-free clostridial clusters or B. fragilis might be better served
mice with B. infantis could suppress the stress response184 by administering probiotics or FMT that are selectively
and now a randomized, placebo-controlled trial has supplemented with these bacteria. Clinical trials stratified
demonstrated symptom improvement and favourable by these parameters are eagerly awaited by the scientific
modulation of mucosal immune responses following and clinical communities.
administration of B. infantis in patients with IBS211.
Nonabsorbable antibiotics offer the opportunity for Conclusions
intestinal microbial modulation without the possibility In conclusion, lines of communication between the brain
of systemic adverse effects. A landmark phase III trial and gut play a key part in the biology, clinical manifes-
reporting the outcome of two separate studies with iden- tations and clinical outcomes of gut diseases, but most
tical protocols (TARGET1 and TARGET2) has shown notably in FGIDs. The central pathways through which
the efficacy of a 2‑week course of rifaximin in diarrhoea-­ this dialogue is mediated are neural and immune net-
predominant IBS, including reduced abdominal pain and works, including crosstalk between these systems. The
reduced diarrhoea episode frequency 212. Dietary modifi­ bidirectional trafficking of these signals opens the pos-
cation is now also a mainstay of IBS treatment. A diet sibility of some gut diseases being driven by aberrant
low in fermentable, oligosaccharides, disaccharides, brain function, and conversely, disordered gut homeo-
monosaccharides and polyols (FODMAPs) substan- stasis being responsible for driving brain pathology and,
tially altered the composition of the intestinal micro- particularly, mood disorders in other patients. Overall,
biota in patients with IBS213, and is efficacious for IBS the mucosal immune system is potentially the key gate-
symptoms213,214. Evidence also exists that low FODMAP keeper of these pathways as it specifically interacts with
diets modulate host metabolic and immune pathways, nerves, the luminal microbiota and is an important tar-
including an eightfold reduction in urinary histamine get of the stress response. What effect these insights will
concentration in patients with IBS consuming a low have on clinical practice remains to be seen, although it
FODMAP diet215. is conceptually appealing to envisage the emergence of
Looking forward, smarter selection and focused new diagnostic criteria, biomarkers and psychological
manipulation of the composition of the microbiota with scoring tools in patients with gut diseases. With the emer-
probiotics or FMT might be required and a manufactured gence of precision medicine, strategies in which patients
‘therapeutic bacterial community’ might comprise high with FGID or IBD might be stratified beyond standard
levels of diversity, depletion of pro-inflammatory species gut symptom complexes might offer the opportunity to
(known to adhere to the epithelium) and enrichment of ­tailor therapies to individual patients. In particular, anti-­
immunosuppressive bacteria. Indeed, supplementation cytokine therapy or efforts to manipulate the intestinal
of bacteria favouring anti-inflammatory IL‑10 responses, microbiota could hold the key to effectively uncoupling
such as B. fragilis and particular clusters of Clostridia, pathological brain–gut dialogue, with the potential to
might hold particular promise to limit excessive mucosal disrupt the proximal drivers of these important diseases.

1. Molodecky, N. A. et al. Increasing incidence and 5. Jostins, L. et al. Host–microbe interactions have 8. Tansey, E. M. Pavlov at home and abroad: his role
prevalence of the inflammatory bowel diseases with shaped the genetic architecture of inflammatory in international physiology. Auton. Neurosci. 125,
time, based on systematic review. Gastroenterology bowel disease. Nature 491, 119–124 (2012). 1–11 (2006).
142, 46–54.e42 (2012). 6. Trynka, G. et al. Dense genotyping identifies 9. Furness, J. B. The enteric nervous system and
2. Talley, N. J. & Ford, A. C. Functional dyspepsia. and localizes multiple common and rare variant neurogastroenterology. Nat. Rev. Gastroenterol.
N. Engl. J. Med. 373, 1853–1863 (2015). association signals in celiac disease. Nat. Genet. Hepatol. 9, 286–294 (2012).
3. Talley, N. J. Functional gastrointestinal disorders as 43, 1193–1201 (2011). 10. Goyal, R. K. & Hirano, I. The enteric nervous system.
a public health problem. Neurogastroenterol. Motil. 7. Ek, W. E. et al. Exploring the genetics of irritable N. Engl. J. Med. 334, 1106–1115 (1996).
20 (Suppl. 1), 121–129 (2008). bowel syndrome: a GWA study in the general 11. Savidge, T. C. et al. Enteric glia regulate intestinal
4. Talley, N. J. & Spiller, R. Irritable bowel syndrome: population and replication in multinational barrier function and inflammation via release
a little understood organic bowel disease? Lancet case–control cohorts. Gut 64, 1774–1782 of S‑nitrosoglutathione. Gastroenterology 132,
360, 555–564 (2002). (2015). 1344–1358 (2007).

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

12. Bohorquez, D. V. & Liddle, R. A. The gut connectome: factor modulate submucosal plexus activity and colonic 56. Drossman, D. A. et al. Sexual and physical abuse
making sense of what you eat. J. Clin. Invest. 125, secretion. Brain Behav. Immun. 30, 115–124 (2013). in women with functional or organic gastrointestinal
888–890 (2015). 35. Reber, S. O., Obermeier, F., Straub, R. H., Falk, W. disorders. Ann. Intern. Med. 113, 828–833 (1990).
13. Mace, O. J., Tehan, B. & Marshall, F. Pharmacology & Neumann, I. D. Chronic intermittent psychosocial 57. Koloski, N. A., Jones, M. & Talley, N. J. Evidence that
and physiology of gastrointestinal enteroendocrine stress (social defeat/overcrowding) in mice increases independent gut‑to‑brain and brain‑to‑gut pathways
cells. Pharmacol. Res. Perspect. 3, e00155 (2015). the severity of an acute DSS-induced colitis operate in the irritable bowel syndrome and functional
14. Mace, O. J. & Marshall, F. Digestive physiology of the and impairs regeneration. Endocrinology 147, dyspepsia: a 1‑year population-based prospective
pig symposium: gut chemosensing and the regulation 4968–4976 (2006). study. Aliment. Pharmacol. Ther. 44, 592–600
of nutrient absorption and energy supply. J. Anim. Sci. 36. Lennon, E. M. et al. Early life stress triggers persistent (2016).
91, 1932–1945 (2013). colonic barrier dysfunction and exacerbates colitis in 58. Thabane, M., Kottachchi, D. T. & Marshall, J. K.
15. Hart, B. L. Biological basis of the behavior of sick adult IL‑10−/− mice. Inflamm. Bowel Dis. 19, 712–719 Systematic review and meta-analysis: the incidence
animals. Neurosci. Biobehav. Rev. 12, 123–137 (2013). and prognosis of post-infectious irritable bowel
(1988). 37. Varghese, A. K. et al. Antidepressants attenuate syndrome. Aliment. Pharmacol. Ther. 26, 535–544
16. Dantzer, R., O’Connor, J. C., Freund, G. G., increased susceptibility to colitis in a murine model (2007).
Johnson, R. W. & Kelley, K. W. From inflammation of depression. Gastroenterology 130, 1743–1753 59. Marshall, J. K. et al. Incidence and epidemiology of
to sickness and depression: when the immune system (2006). irritable bowel syndrome after a large waterborne
subjugates the brain. Nat. Rev. Neurosci. 9, 46–56 38. Qiu, B. S., Vallance, B. A., Blennerhassett, P. A. outbreak of bacterial dysentery. Gastroenterology
(2008). & Collins, S. M. The role of CD4+ lymphocytes in 131, 445–450 (2006).
17. Okada, H., Kuhn, C., Feillet, H. & Bach, J. F. The ‘hygiene the susceptibility of mice to stress-induced reactivation 60. Ford, A. C. et al. Prevalence of uninvestigated
hypothesis’ for autoimmune and allergic diseases: of experimental colitis. Nat. Med. 5, 1178–1182 dyspepsia 8 years after a large waterborne
an update. Clin. Exp. Immunol. 160, 1–9 (2010). (1999). outbreak of bacterial dysentery: a cohort study.
18. Glaser, R., Robles, T. F., Sheridan, J., Malarkey, W. B. 39. Adam, B. et al. Severity of mucosal inflammation as Gastroenterology 138, 1727–1736 (2010).
& Kiecolt-Glaser, J. K. Mild depressive symptoms are a predictor for alterations of visceral sensory function 61. Wouters, M. M. et al. Psychological comorbidity
associated with amplified and prolonged inflammatory in a rat model. Pain 123, 179–186 (2006). increases the risk for postinfectious IBS partly
responses after influenza virus vaccination in older 40. Mykletun, A. et al. Prevalence of mood and anxiety by enhanced susceptibility to develop infectious
adults. Arch. Gen. Psychiatry 60, 1009–1014 (2003). disorder in self reported irritable bowel syndrome gastroenteritis. Gut 65, 1279–1288 (2016).
19. McEwen, B. S. Stress, adaptation, and disease: (IBS). An epidemiological population based study 62. Dunlop, S. P., Jenkins, D., Neal, K. R. & Spiller, R. C.
allostasis and allostatic load. Ann. NY Acad. Sci. 840, of women. BMC Gastroenterol. 10, 88 (2010). Relative importance of enterochromaffin cell
33–44 (1998). 41. Koloski, N. A. et al. The brain–gut pathway in hyperplasia, anxiety, and depression in postinfectious
20. Goodhand, J. R. et al. Mood disorders in inflammatory functional gastrointestinal disorders is bidirectional: IBS. Gastroenterology 125, 1651–1659 (2003).
bowel disease: relation to diagnosis, disease activity, a 12‑year prospective population-based study. Gut 61, 63. Neurath, M. F. Cytokines in inflammatory bowel
perceived stress, and other factors. Inflamm. Bowel Dis. 1284–1290 (2012). disease. Nat. Rev. Immunol. 14, 329–342 (2014).
18, 2301–2309 (2012). 42. Savas, L. S. et al. Irritable bowel syndrome and 64. Isidro, R. A. & Appleyard, C. B. Colonic macrophage
21. Addolorato, G., Capristo, E., Stefanini, G. F. dyspepsia among women veterans: prevalence polarization in homeostasis, inflammation, and cancer.
& Gasbarrini, G. Inflammatory bowel disease: a study and association with psychological distress. Am. J. Physiol. Gastrointest. Liver Physiol. 311,
of the association between anxiety and depression, Aliment. Pharmacol. Ther. 29, 115–125 (2009). G59–G73 (2016).
physical morbidity, and nutritional status. 43. Jones, R., Latinovic, R., Charlton, J. & Gulliford, M. 65. La Flamme, A. C. et al. Type II‑activated murine
Scand. J. Gastroenterol. 32, 1013–1021 (1997). Physical and psychological co‑morbidity in irritable macrophages produce IL‑4. PLoS ONE 7, e46989
22. Kovacs, Z. & Kovacs, F. Depressive and anxiety bowel syndrome: a matched cohort study using (2012).
symptoms, dysfunctional attitudes and social aspects the General Practice Research Database. 66. Goldberg, R., Prescott, N., Lord, G. M.,
in irritable bowel syndrome and inflammatory bowel Aliment. Pharmacol. Ther. 24, 879–886 (2006). MacDonald, T. T. & Powell, N. The unusual suspects
disease. Int. J. Psychiatry Med. 37, 245–255 (2007). 44. Tayama, J. et al. Maladjustment to academic life — innate lymphoid cells as novel therapeutic targets
23. Kurina, L. M., Goldacre, M. J., Yeates, D. & Gill, L. E. and employment anxiety in university students with in IBD. Nat. Rev. Gastroenterol. Hepatol. 12,
Depression and anxiety in people with inflammatory irritable bowel syndrome. PLoS ONE 10, e0129345 271–283 (2015).
bowel disease. J. Epidemiol. Community Health 55, (2015). 67. Goto, Y. et al. Innate lymphoid cells regulate intestinal
716–720 (2001). 45. Vu, J., Kushnir, V., Cassell, B., Gyawali, C. P. epithelial cell glycosylation. Science 345, 1254009
24. Hauser, W., Janke, K. H., Klump, B. & Hinz, A. & Sayuk, G. S. The impact of psychiatric and (2014).
Anxiety and depression in patients with inflammatory extraintestinal comorbidity on quality of life and bowel 68. Zimmerman, N. P., Vongsa, R. A., Wendt, M. K.
bowel disease: comparisons with chronic liver disease symptom burden in functional GI disorders. & Dwinell, M. B. Chemokines and chemokine
patients and the general population. Inflamm. Bowel Neurogastroenterol. Motil. 26, 1323–1332 (2014). receptors in mucosal homeostasis at the intestinal
Dis. 17, 621–632 (2011). 46. Kennedy, P. J. et al. Cognitive performance in irritable epithelial barrier in inflammatory bowel disease.
25. Mardini, H. E., Kip, K. E. & Wilson, J. W. Crohn’s bowel syndrome: evidence of a stress-related Inflamm. Bowel Dis. 14, 1000–1011 (2008).
disease: a two-year prospective study of the impairment in visuospatial memory. Psychol. Med. 69. Charo, I. F. & Ransohoff, R. M. The many roles of
association between psychological distress and 44, 1553–1566 (2014). chemokines and chemokine receptors in inflammation.
disease activity. Dig. Dis. Sci. 49, 492–497 (2004). 47. Canavan, J. B., Bennett, K., Feely, J., O’Morain, C. A. N. Engl. J. Med. 354, 610–621 (2006).
26. Persoons, P. et al. The impact of major depressive & O’Connor, H. J. Significant psychological morbidity 70. Habtezion, A., Nguyen, L. P., Hadeiba, H.
disorder on the short- and long-term outcome occurs in irritable bowel syndrome: a case–control & Butcher, E. C. Leukocyte trafficking to the small
of Crohn’s disease treatment with infliximab. study using a pharmacy reimbursement database. intestine and colon. Gastroenterology 150, 340–354
Aliment. Pharmacol. Ther. 22, 101–110 (2005). Aliment. Pharmacol. Ther. 29, 440–449 (2009). (2016).
27. Mittermaier, C. et al. Impact of depressive mood on 48. Qi, R. et al. Intrinsic brain abnormalities in irritable 71. Feagan, B. G. et al. Vedolizumab as induction and
relapse in patients with inflammatory bowel disease: bowel syndrome and effect of anxiety and depression. maintenance therapy for ulcerative colitis. N. Engl.
a prospective 18‑month follow‑up study. Brain Imaging Behav. http://dx.doi.org/10.1007/ J. Med. 369, 699–710 (2013).
Psychosom. Med. 66, 79–84 (2004). s11682-015-9478-1 (2015). 72. Sandborn, W. J. et al. Vedolizumab as induction
28. Bernstein, C. N. et al. A prospective population-based 49. Icenhour, A. et al. Neural circuitry of abdominal and maintenance therapy for Crohn’s disease. N. Engl.
study of triggers of symptomatic flares in IBD. pain‑related fear learning and reinstatement in J. Med. 369, 711–721 (2013).
Am. J. Gastroenterol. 105, 1994–2002 (2010). irritable bowel syndrome. Neurogastroenterol. Motil. 73. Ulrich-Lai, Y. M. & Herman, J. P. Neural regulation of
29. Goodhand, J. R. et al. Factors associated with 27, 114–127 (2015). endocrine and autonomic stress responses. Nat. Rev.
thiopurine non-adherence in patients with 50. Song, G. H. et al. Cortical effects of anticipation Neurosci. 10, 397–409 (2009).
inflammatory bowel disease. Aliment. Pharmacol. and endogenous modulation of visceral pain assessed 74. Elenkov, I. J., Wilder, R. L., Chrousos, G. P. & Vizi, E. S.
Ther. 38, 1097–1108 (2013). by functional brain MRI in irritable bowel syndrome The sympathetic nerve — an integrative interface
30. Larauche, M., Mulak, A. & Tache, Y. Stress and visceral patients and healthy controls. Pain 126, 79–90 between two supersystems: the brain and the immune
pain: from animal models to clinical therapies. (2006). system. Pharmacol. Rev. 52, 595–638 (2000).
Exp. Neurol. 233, 49–67 (2012). 51. Hong, J. Y. et al. Altered brain responses in subjects 75. Felten, D. L., Felten, S. Y., Carlson, S. L.,
31. Larauche, M., Gourcerol, G., Million, M., with irritable bowel syndrome during cued and uncued Olschowka, J. A. & Livnat, S. Noradrenergic and
Adelson, D. W. & Tache, Y. Repeated psychological pain expectation. Neurogastroenterol. Motil. 28, peptidergic innervation of lymphoid tissue. J. Immunol.
stress-induced alterations of visceral sensitivity and 127–138 (2016). 135, 755s–765s (1985).
colonic motor functions in mice: influence of surgery 52. Grzesiak, M. et al. The lifetime prevalence of anxiety 76. Mawdsley, J. E., Macey, M. G., Feakins, R. M.,
and postoperative single housing on visceromotor disorders among patients with irritable bowel Langmead, L. & Rampton, D. S. The effect of acute
responses. Stress 13, 343–354 (2010). syndrome. Adv. Clin. Exp. Med. 23, 987–992 (2014). psychologic stress on systemic and rectal mucosal
32. Bradesi, S., Eutamene, H., Garcia-Villar, R., 53. Hillila, M. T., Siivola, M. T. & Farkkila, M. A. measures of inflammation in ulcerative colitis.
Fioramonti, J. & Bueno, L. Acute and chronic stress Comorbidity and use of health-care services among Gastroenterology 131, 410–419 (2006).
differently affect visceral sensitivity to rectal distension irritable bowel syndrome sufferers. Scand. J. 77. Vanuytsel, T. et al. Psychological stress and
in female rats. Neurogastroenterol. Motil. 14, 75–82 Gastroenterol. 42, 799–806 (2007). corticotropin-releasing hormone increase intestinal
(2002). 54. Talley, N. J., Fett, S. L., Zinsmeister, A. R. permeability in humans by a mast cell-dependent
33. Ren, T. H. et al. Effects of neonatal maternal & Melton, L. J. 3rd. Gastrointestinal tract symptoms mechanism. Gut 63, 1293–1299 (2014).
separation on neurochemical and sensory response and self-reported abuse: a population-based study. 78. Nakano, K. et al. Dopamine induces IL‑6‑dependent
to colonic distension in a rat model of irritable bowel Gastroenterology 107, 1040–1049 (1994). IL‑17 production via D1‑like receptor on CD4 naive
syndrome. Am. J. Physiol. Gastrointest. Liver Physiol. 55. Talley, N. J., Fett, S. L. & Zinsmeister, A. R. T cells and D1‑like receptor antagonist SCH‑23390
292, G849–G856 (2007). Self‑reported abuse and gastrointestinal disease inhibits cartilage destruction in a human rheumatoid
34. O’Malley, D., Cryan, J. F. & Dinan, T. G. Crosstalk in outpatients: association with irritable bowel-type arthritis/SCID mouse chimera model. J. Immunol.
between interleukin‑6 and corticotropin-releasing symptoms. Am. J. Gastroenterol. 90, 366–371 (1995). 186, 3745–3752 (2011).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 15


©
2
0
1
7
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
.
REVIEWS

79. Khan, N. A. & Poisson, J. P. 5‑HT3 receptor-channels 103. Bashashati, M. et al. Cytokine imbalance in irritable functional disorders. Nat. Rev. Gastroenterol. Hepatol.
coupled with Na+ influx in human T cells: role in T cell bowel syndrome: a systematic review and meta- 7, 146–156 (2010).
activation. J. Neuroimmunol. 99, 53–60 (1999). analysis. Neurogastroenterol. Motil. 26, 1036–1048 127. Kindt, S. et al. Immune dysfunction in patients
80. Aune, T. M., McGrath, K. M., Sarr, T., Bombara, M. P. (2014). with functional gastrointestinal disorders.
& Kelley, K. A. Expression of 5HT1a receptors on 104. Dinan, T. G. et al. Hypothalamic–pituitary–gut axis Neurogastroenterol. Motil. 21, 389–398 (2009).
activated human T cells. Regulation of cyclic AMP dysregulation in irritable bowel syndrome: plasma 128. Ohman, L., Isaksson, S., Lundgren, A., Simren, M.
levels and T cell proliferation by 5‑hydroxytryptamine. cytokines as a potential biomarker? Gastroenterology & Sjovall, H. A controlled study of colonic immune
J. Immunol. 151, 1175–1183 (1993). 130, 304–311 (2006). activity and β7+ blood T lymphocytes in patients with
81. Buttari, B. et al. Neuropeptide Y induces potent 105. Dinan, T. G. et al. Enhanced cholinergic-mediated irritable bowel syndrome. Clin. Gastroenterol. Hepatol.
migration of human immature dendritic cells increase in the pro-inflammatory cytokine IL‑6 3, 980–986 (2005).
and promotes a Th2 polarization. FASEB J. 28, in irritable bowel syndrome: role of muscarinic 129. Spiller, R. C. et al. Increased rectal mucosal
3038–3049 (2014). receptors. Am. J. Gastroenterol. 103, 2570–2576 enteroendocrine cells, T lymphocytes, and increased
82. Payan, D. G., Brewster, D. R., Missirian-Bastian, A. (2008). gut permeability following acute Campylobacter
& Goetzl, E. J. Substance P recognition by a subset of 106. Liebregts, T. et al. Immune activation in patients enteritis and in post-dysenteric irritable bowel
human T lymphocytes. J. Clin. Invest. 74, 1532–1539 with irritable bowel syndrome. Gastroenterology 132, syndrome. Gut 47, 804–811 (2000).
(1984). 913–920 (2007). 130. Elsenbruch, S. et al. Are there alterations of
83. Johnson, M. C., McCormack, R. J., Delgado, M., 107. Liebregts, T. et al. Small bowel homing T cells neuroendocrine and cellular immune responses
Martinez, C. & Ganea, D. Murine T‑lymphocytes are associated with symptoms and delayed to nutrients in women with irritable bowel syndrome?
express vasoactive intestinal peptide receptor 1 gastric emptying in functional dyspepsia. Am. J. Am. J. Gastroenterol. 99, 703–710 (2004).
(VIP‑R1) mRNA. J. Neuroimmunol. 68, 109–119 Gastroenterol. 106, 1089–1098 (2011). 131. Bal, S. M. et al. IL‑1β, IL‑4 and IL‑12 control the
(1996). 108. Gao, J. Correlation between anxiety-depression status fate of group 2 innate lymphoid cells in human
84. Sanders, V. M. et al. Differential expression of the and cytokines in diarrhea-predominant irritable bowel airway inflammation in the lungs. Nat. Immunol. 17,
β2‑adrenergic receptor by Th1 and Th2 clones: syndrome. Exp. Ther. Med. 6, 93–96 (2013). 636–645 (2016).
implications for cytokine production and B cell help. 109. Aerssens, J. et al. Alterations in mucosal immunity 132. Suto, G., Kiraly, A. & Tache, Y. Interleukin 1β inhibits
J. Immunol. 158, 4200–4210 (1997). identified in the colon of patients with irritable bowel gastric emptying in rats: mediation through
85. Sanders, V. M. The role of adrenoceptor-mediated syndrome. Clin. Gastroenterol. Hepatol. 6, 194–205 prostaglandin and corticotropin-releasing factor.
signals in the modulation of lymphocyte function. (2008). Gastroenterology 106, 1568–1575 (1994).
Adv. Neuroimmunol. 5, 283–298 (1995). 110. Macsharry, J. et al. Mucosal cytokine imbalance in 133. Hermann, G. & Rogers, R. C. Tumor necrosis factor-
86. Sanders, V. M. & Straub, R. H. Norepinephrine, the irritable bowel syndrome. Scand. J. Gastroenterol. 43, alpha in the dorsal vagal complex suppresses gastric
β-adrenergic receptor, and immunity. Brain Behav. 1467–1476 (2008). motility. Neuroimmunomodulation 2, 74–81 (1995).
Immun. 16, 290–332 (2002). 111. Barbara, G. et al. Activated mast cells in proximity 134. Benakis, C. et al. Commensal microbiota affects
87. Panina-Bordignon, P. et al. β2‑agonists prevent Th1 to colonic nerves correlate with abdominal pain in ischemic stroke outcome by regulating intestinal
development by selective inhibition of interleukin 12. irritable bowel syndrome. Gastroenterology 126, γδ T cells. Nat. Med. 22, 516–523 (2016).
J. Clin. Invest. 100, 1513–1519 (1997). 693–702 (2004). 135. Felger, J. C. & Lotrich, F. E. Inflammatory cytokines
88. Ramer-Quinn, D. S., Baker, R. A. & Sanders, V. M. 112. Guilarte, M. et al. Diarrhoea-predominant IBS patients in depression: neurobiological mechanisms and
Activated T helper 1 and T helper 2 cells differentially show mast cell activation and hyperplasia in the therapeutic implications. Neuroscience 246,
express the beta‑2‑adrenergic receptor: a mechanism jejunum. Gut 56, 203–209 (2007). 199–229 (2013).
for selective modulation of T helper 1 cell cytokine 113. Martinez, C. et al. Diarrhoea-predominant irritable 136. Emch, G. S., Hermann, G. E. & Rogers, R. C.
production. J. Immunol. 159, 4857–4867 (1997). bowel syndrome: an organic disorder with structural TNF-α activates solitary nucleus neurons responsive
89. Takenaka, M. C. et al. Norepinephrine controls effector abnormalities in the jejunal epithelial barrier. Gut 62, to gastric distension. Am. J. Physiol. Gastrointest.
T cell differentiation through β2‑adrenergic receptor- 1160–1168 (2013). Liver Physiol. 279, G582–G586 (2000).
mediated inhibition of NF‑κB and AP‑1 in dendritic 114. Weston, A. P., Biddle, W. L., Bhatia, P. S. 137. Soulet, D. & Rivest, S. Bone-marrow-derived microglia:
cells. J. Immunol. 196, 637–644 (2016). & Miner, P. B. Jr. Terminal ileal mucosal mast cells myth or reality? Curr. Opin. Pharmacol. 8, 508–518
90. Orand, A. et al. Catecholaminergic gene in irritable bowel syndrome. Dig. Dis. Sci. 38, (2008).
polymorphisms are associated with GI symptoms 1590–1595 (1993). 138. Rivest, S. Molecular insights on the cerebral innate
and morphological brain changes in irritable bowel 115. Akbar, A. et al. Increased capsaicin receptor immune system. Brain Behav. Immun. 17, 13–19
syndrome. PLoS ONE 10, e0135910 (2015). TRPV1‑expressing sensory fibres in irritable bowel (2003).
91. Gabanyi, I. et al. Neuro-immune interactions drive syndrome and their correlation with abdominal pain. 139. Jack, C. S. et al. TLR signaling tailors innate immune
tissue programming in intestinal macrophages. Cell Gut 57, 923–929 (2008). responses in human microglia and astrocytes.
164, 378–391 (2016). 116. Cremon, C. et al. Mucosal immune activation in J. Immunol. 175, 4320–4330 (2005).
92. Kurowska-Stolarska, M. et al. IL‑33 amplifies the irritable bowel syndrome: gender-dependence 140. Arnett, H. A. et al. TNFα promotes proliferation
polarization of alternatively activated macrophages and association with digestive symptoms. Am. J. of oligodendrocyte progenitors and remyelination.
that contribute to airway inflammation. J. Immunol. Gastroenterol. 104, 392–400 (2009). Nat. Neurosci. 4, 1116–1122 (2001).
183, 6469–6477 (2009). 117. Barbara, G. et al. Mast cell-dependent excitation of 141. Klein, M. et al. Innate immunity to pneumococcal
93. Li, D. et al. IL‑33 promotes ST2‑dependent lung fibrosis visceral-nociceptive sensory neurons in irritable bowel infection of the central nervous system depends on
by the induction of alternatively activated macrophages syndrome. Gastroenterology 132, 26–37 (2007). Toll-like receptor (TLR) 2 and TLR4. J. Infect. Dis. 198,
and innate lymphoid cells in mice. J. Allergy Clin. 118. Buhner, S. et al. Submucous rather than myenteric 1028–1036 (2008).
Immunol. 134, 1422–1432.e11 (2014). neurons are activated by mucosal biopsy supernatants 142. Zhang, S. Y. et al. TLR3 deficiency in patients
94. Cirillo, C. et al. Evidence for neuronal and structural from irritable bowel syndrome patients. with herpes simplex encephalitis. Science 317,
changes in submucous ganglia of patients with Neurogastroenterol. Motil. 24, 1134–e572 (2012). 1522–1527 (2007).
functional dyspepsia. Am. J. Gastroenterol. 110, 119. Wouters, M. M. et al. Histamine receptor H1‑mediated 143. Bogie, J. F., Stinissen, P. & Hendriks, J. J.
1205–1215 (2015). sensitization of TRPV1 mediates visceral Macrophage subsets and microglia in multiple
95. Girodet, P. O. et al. Alternative macrophage activation hypersensitivity and symptoms in patients with sclerosis. Acta Neuropathol. 128, 191–213 (2014).
is increased in asthma. Am. J. Respir. Cell Mol. Biol. irritable bowel syndrome. Gastroenterology 150, 144. Liu, Y. et al. LPS receptor (CD14): a receptor for
55, 467–475 (2016). 875–887. e89 (2016). phagocytosis of Alzheimer’s amyloid peptide. Brain
96. Airaksinen, M. S. & Saarma, M. The GDNF family: 120. Santos, J., Yang, P. C., Soderholm, J. D., Benjamin, M. 128, 1778–1789 (2005).
signalling, biological functions and therapeutic value. & Perdue, M. H. Role of mast cells in chronic stress 145. Casellas, F., Aguade, S. & Molero, J. Intestinal
Nat. Rev. Neurosci. 3, 383–394 (2002). induced colonic epithelial barrier dysfunction in permeability in inflammatory bowel disease. Am. J.
97. Ibiza, S. et al. Glial-cell-derived neuroregulators the rat. Gut 48, 630–636 (2001). Gastroenterol. 81, 502 (1986).
control type 3 innate lymphoid cells and gut defence. 121. Ronkainen, J. et al. Prevalence of oesophageal 146. Mujagic, Z. et al. Small intestinal permeability is
Nature 535, 440–443 (2016). eosinophils and eosinophilic oesophagitis in adults: increased in diarrhoea predominant IBS, while
98. Limsui, D. et al. Symptomatic overlap between the population-based Kalixanda study. Gut 56, alterations in gastroduodenal permeability in all IBS
irritable bowel syndrome and microscopic colitis. 615–620 (2007). subtypes are largely attributable to confounders.
Inflamm. Bowel Dis. 13, 175–181 (2007). 122. Vanheel, H. et al. Impaired duodenal mucosal integrity Aliment. Pharmacol. Ther. 40, 288–297 (2014).
99. Midhagen, G. & Hallert, C. High rate of and low-grade inflammation in functional dyspepsia. 147. Dlugosz, A. et al. Increased serum levels of
gastrointestinal symptoms in celiac patients living Gut 63, 262–271 (2014). lipopolysaccharide and antiflagellin antibodies
on a gluten-free diet: controlled study. Am. J. 123. Walker, M. M. et al. Duodenal eosinophilia and early in patients with diarrhea-predominant irritable
Gastroenterol. 98, 2023–2026 (2003). satiety in functional dyspepsia: confirmation of bowel syndrome. Neurogastroenterol. Motil. 27,
100. Isgar, B., Harman, M., Kaye, M. D. & Whorwell, P. J. a positive association in an Australian cohort. 1747–1754 (2015).
Symptoms of irritable bowel syndrome in ulcerative J. Gastroenterol. Hepatol. 29, 474–479 (2014). 148. McDonnell, M. et al. Systemic Toll-like receptor
colitis in remission. Gut 24, 190–192 (1983). 124. Walker, M. M. et al. Implications of eosinophilia in ligands modify B‑cell responses in human
101. Farrokhyar, F., Marshall, J. K., Easterbrook, B. the normal duodenal biopsy — an association with inflammatory bowel disease. Inflamm. Bowel Dis. 17,
& Irvine, E. J. Functional gastrointestinal disorders allergy and functional dyspepsia. Aliment. Pharmacol. 298–307 (2011).
and mood disorders in patients with inactive Ther. 31, 1229–1236 (2010). 149. Dunn, A. J., Swiergiel, A. H. & de Beaurepaire, R.
inflammatory bowel disease: prevalence and impact 125. Futagami, S. et al. Migration of eosinophils and Cytokines as mediators of depression: what can we
on health. Inflamm. Bowel Dis. 12, 38–46 (2006). CCR2-/CD68‑double positive cells into the duodenal learn from animal studies? Neurosci. Biobehav. Rev.
102. Ford, A. C., Talley, N. J., Walker, M. M. & Jones, M. P. mucosa of patients with postinfectious functional 29, 891–909 (2005).
Increased prevalence of autoimmune diseases in dyspepsia. Am. J. Gastroenterol. 105, 1835–1842 150. Kronfol, Z. & House, J. D. Lymphocyte mitogenesis,
functional gastrointestinal disorders: case–control (2010). immunoglobulin and complement levels in depressed
study of 23471 primary care patients. 126. Powell, N., Walker, M. M. & Talley, N. J. patients and normal controls. Acta Psychiatr. Scand.
Aliment. Pharmacol. Ther. 40, 827–834 (2014). Gastrointestinal eosinophils in health, disease and 80, 142–147 (1989).

16 | ADVANCE ONLINE PUBLICATION www.nature.com/nrgastro


©
2
0
1
7
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
.
REVIEWS

151. Maes, M. et al. Evidence for a systemic immune 174. Souza, D. G. et al. The essential role of the intestinal in irritable bowel syndrome: systematic review
activation during depression: results of leukocyte microbiota in facilitating acute inflammatory and meta-analysis. Am. J. Gastroenterol. 109,
enumeration by flow cytometry in conjunction with responses. J. Immunol. 173, 4137–4146 (2004). 1350–1365 (2014).
monoclonal antibody staining. Psychol. Med. 22, 175. Ivanov, I. I. et al. Induction of intestinal Th17 cells by 199. Lackner, J. M. et al. Rapid response to cognitive
45–53 (1992). segmented filamentous bacteria. Cell 139, 485–498 behavior therapy predicts treatment outcome
152. Miyaoka, H. et al. Depression from interferon therapy (2009). in patients with irritable bowel syndrome.
in patients with hepatitis C. Am. J. Psychiatry 156, 176. Atarashi, K. et al. Th17 cell induction by adhesion Clin. Gastroenterol. Hepatol. 8, 426–432 (2010).
1120 (1999). of microbes to intestinal epithelial cells. Cell 163, 200. Everitt, H. et al. Assessing Cognitive behavioural
153. Loftis, J. M. et al. Vulnerability to somatic symptoms 367–380 (2015). Therapy in Irritable Bowel (ACTIB): protocol for a
of depression during interferon-alpha therapy 177. Seo, S. U. et al. Distinct commensals induce randomised controlled trial of clinical-effectiveness
for hepatitis C: a 16‑week prospective study. interleukin‑1β via NLRP3 inflammasome in and cost-effectiveness of therapist delivered cognitive
J. Psychosom. Res. 74, 57–63 (2013). inflammatory monocytes to promote intestinal behavioural therapy and web-based self-management
154. Dantzer, R. & Kelley, K. W. Twenty years of research inflammation in response to injury. Immunity 42, in irritable bowel syndrome in adults. BMJ Open 5,
on cytokine-induced sickness behavior. Brain Behav. 744–755 (2015). e008622 (2015).
Immun. 21, 153–160 (2007). 178. Lakhdari, O. et al. Functional metagenomics: a high 201. Lackner, J. M. et al. The Irritable Bowel Syndrome
155. Kapas, L. et al. Somnogenic, pyrogenic, and anorectic throughput screening method to decipher microbiota- Outcome Study (IBSOS): rationale and design of a
activities of tumor necrosis factor-alpha and TNF- driven NF‑κB modulation in the human gut. PLoS ONE randomized, placebo-controlled trial with 12 month
alpha fragments. Am. J. Physiol. 263, R708–R715 5, e13092 (2010). follow up of self- versus clinician-administered CBT
(1992). 179. Powell, N. et al. The transcription factor T‑bet for moderate to severe irritable bowel syndrome.
156. Kent, S., Rodriguez, F., Kelley, K. W. & Dantzer, R. regulates intestinal inflammation mediated by Contemp. Clin. Trials 33, 1293–1310 (2012).
Reduction in food and water intake induced interleukin‑7 receptor+ innate lymphoid cells. 202. Dunlop, S. P. et al. Randomized, double-blind,
by microinjection of interleukin‑1 beta in the Immunity 37, 674–684 (2012). placebo-controlled trial of prednisolone in
ventromedial hypothalamus of the rat. Physiol. Behav. 180. Furusawa, Y. et al. Commensal microbe-derived post‑infectious irritable bowel syndrome.
56, 1031–1036 (1994). butyrate induces the differentiation of colonic Aliment. Pharmacol. Ther. 18, 77–84 (2003).
157. Besedovsky, H., del Rey, A., Sorkin, E. regulatory T cells. Nature 504, 446–450 (2013). 203. Feldmann, M. Many cytokines are very useful
& Dinarello, C. A. Immunoregulatory feedback 181. Smith, P. M. et al. The microbial metabolites, short- therapeutic targets in disease. J. Clin. Invest. 118,
between interleukin‑1 and glucocorticoid hormones. chain fatty acids, regulate colonic Treg cell homeostasis. 3533–3536 (2008).
Science 233, 652–654 (1986). Science 341, 569–573 (2013). 204. Stefanini, G. F. et al. Oral cromolyn sodium in
158. Silverman, M. N., Pearce, B. D. & Miller, A. H. 182. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ comparison with elimination diet in the irritable bowel
in Cytokines and Mental Health (ed Kronfol, Z.) regulatory T‑cell development by a commensal syndrome, diarrheic type. Multicenter study of 428
85–122 (Springer Science, 2003). bacterium of the intestinal microbiota. Proc. Natl patients. Scand. J. Gastroenterol. 30, 535–541
159. Mark, K. S. & Miller, D. W. Increased permeability Acad. Sci. USA 107, 12204–12209 (2010). (1995).
of primary cultured brain microvessel endothelial cell 183. Bailey, M. T. et al. Exposure to a social stressor alters 205. Lunardi, C. et al. Double-blind cross-over trial of oral
monolayers following TNF-α exposure. Life Sci. 64, the structure of the intestinal microbiota: implications sodium cromoglycate in patients with irritable bowel
1941–1953 (1999). for stressor-induced immunomodulation. Brain Behav. syndrome due to food intolerance. Clin. Exp. Allergy
160. Lopez-Ramirez, M. A. et al. Role of caspases in Immun. 25, 397–407 (2011). 21, 569–572 (1991).
cytokine-induced barrier breakdown in human brain 184. Sudo, N. et al. Postnatal microbial colonization 206. Bahi, A., Schwed, J. S., Walter, M., Stark, H.
endothelial cells. J. Immunol. 189, 3130–3139 programs the hypothalamic–pituitary–adrenal system & Sadek, B. Anxiolytic and antidepressant-like
(2012). for stress response in mice. J. Physiol. 558, 263–275 activities of the novel and potent non-imidazole
161. Cohen, S. S. et al. Effects of interleukin‑6 on the (2004). histamine H3 receptor antagonist ST‑1283. Drug Des.
expression of tight junction proteins in isolated 185. Clarke, G. et al. The microbiome–gut–brain axis Devel. Ther. 8, 627–637 (2014).
cerebral microvessels from yearling and adult sheep. during early life regulates the hippocampal 207. Spiller, R. Review article: probiotics and prebiotics in
Neuroimmunomodulation 20, 264–273 (2013). serotonergic system in a sex-dependent manner. irritable bowel syndrome. Aliment. Pharmacol. Ther.
162. Maruo, N., Morita, I., Shirao, M. & Murota, S. Mol. Psychiatry 18, 666–673 (2013). 28, 385–396 (2008).
IL‑6 increases endothelial permeability in vitro. 186. Neufeld, K. M., Kang, N., Bienenstock, J. 208. Marchesi, J. R. et al. The gut microbiota and host
Endocrinology 131, 710–714 (1992). & Foster, J. A. Reduced anxiety-like behavior and health: a new clinical frontier. Gut 65, 330–339
163. Bsibsi, M. et al. Toll-like receptor 3 on adult human central neurochemical change in germ-free mice. (2016).
astrocytes triggers production of neuroprotective Neurogastroenterol. Motil. 23, 255–e119 (2011). 209. Moran, C. & Shanahan, F. Editorial: probiotics and IBS
mediators. Glia 53, 688–695 (2006). 187. Diaz Heijtz, R. et al. Normal gut microbiota modulates — where are we now? Aliment. Pharmacol. Ther. 40,
164. Human Microbiome Project Consortium. Structure, brain development and behavior. Proc. Natl Acad. 318 (2014).
function and diversity of the healthy human Sci. USA 108, 3047–3052 (2011). 210. Pinn, D. M., Aroniadis, O. C. & Brandt, L. J. Is fecal
microbiome. Nature 486, 207–214 (2012). 188. Galley, J. D. et al. Exposure to a social stressor microbiota transplantation (FMT) an effective
165. Kostic, A. D., Xavier, R. J. & Gevers, D. The disrupts the community structure of the colonic treatment for patients with functional gastrointestinal
microbiome in inflammatory bowel disease: current mucosa-associated microbiota. BMC Microbiol. 14, disorders (FGID)? Neurogastroenterol. Motil. 27,
status and the future ahead. Gastroenterology 146, 189 (2014). 19–29 (2015).
1489–1499 (2014). 189. Galley, J. D. & Bailey, M. T. Impact of stressor exposure 211. O’Mahony, L. et al. Lactobacillus and bifidobacterium
166. Chassaing, B. et al. Crohn disease — associated on the interplay between commensal microbiota and in irritable bowel syndrome: symptom responses and
adherent-invasive E. coli bacteria target mouse host inflammation. Gut Microbes 5, 390–396 (2014). relationship to cytokine profiles. Gastroenterology
and human Peyer’s patches via long polar fimbriae. 190. Galley, J. D. et al. The structures of the colonic 128, 541–551 (2005).
J. Clin. Invest. 121, 966–975 (2011). mucosa-associated and luminal microbial communities 212. Pimentel, M. et al. Rifaximin therapy for patients
167. Sokol, H. et al. Faecalibacterium prausnitzii is an are distinct and differentially affected by a prolonged with irritable bowel syndrome without constipation.
anti‑inflammatory commensal bacterium identified murine stressor. Gut Microbes 5, 748–760 (2014). N. Engl. J. Med. 364, 22–32 (2011).
by gut microbiota analysis of Crohn disease patients. 191. Bailey, M. T. et al. Stressor exposure disrupts 213. Chumpitazi, B. P. et al. Randomised clinical trial: gut
Proc. Natl Acad. Sci. USA 105, 16731–16736 (2008). commensal microbial populations in the intestines microbiome biomarkers are associated with clinical
168. Machiels, K. et al. A decrease of the butyrate- and leads to increased colonization by Citrobacter response to a low FODMAP diet in children with the
producing species Roseburia hominis and rodentium. Infect. Immun. 78, 1509–1519 (2010). irritable bowel syndrome. Aliment. Pharmacol. Ther.
Faecalibacterium prausnitzii defines dysbiosis in 192. Gareau, M. G. et al. Bacterial infection causes 42, 418–427 (2015).
patients with ulcerative colitis. Gut 63, 1275–1283 stress‑induced memory dysfunction in mice. Gut 60, 214. Halmos, E. P., Power, V. A., Shepherd, S. J.,
(2014). 307–317 (2011). Gibson, P. R. & Muir, J. G. A diet low in FODMAPs
169. Carroll, I. M., Ringel-Kulka, T., Siddle, J. P. 193. Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T. G. reduces symptoms of irritable bowel syndrome.
& Ringel, Y. Alterations in composition and diversity & Cryan, J. F. Microbiota is essential for social Gastroenterology 146, 67–75.e5 (2014).
of the intestinal microbiota in patients with development in the mouse. Mol. Psychiatry 19, 215. McIntosh, K. et al. FODMAPs alter symptoms and
diarrhea‑predominant irritable bowel syndrome. 146–148 (2014). the metabolome of patients with IBS: a randomised
Neurogastroenterol. Motil. 24, 521–e248 (2012). 194. Houlden, A. et al. Brain injury induces specific changes controlled trial. Gut http://dx.doi.org/10.1136/
170. Sundin, J. et al. Altered faecal and mucosal microbial in the caecal microbiota of mice via altered autonomic gutjnl-2015-311339 (2016).
composition in post-infectious irritable bowel activity and mucoprotein production. Brain Behav. 216. Zucchelli, M. et al. Association of TNFSF15
syndrome patients correlates with mucosal Immun. 57, 10–20 (2016). polymorphism with irritable bowel syndrome. Gut 60,
lymphocyte phenotypes and psychological distress. 195. Kabouridis, P. S. & Pachnis, V. Emerging roles of gut 1671–1677 (2011).
Aliment. Pharmacol. Ther. 41, 342–351 (2015). microbiota and the immune system in the 217. Swan, C. et al. Identifying and testing candidate
171. Giamarellos-Bourboulis, E. et al. Molecular development of the enteric nervous system. J. Clin. genetic polymorphisms in the irritable bowel
assessment of differences in the duodenal microbiome Invest. 125, 956–964 (2015). syndrome (IBS): association with TNFSF15 and TNFα.
in subjects with irritable bowel syndrome. 196. Collins, J., Borojevic, R., Verdu, E. F., Huizinga, J. D. & Gut 62, 985–994 (2013).
Scand. J. Gastroenterol. 50, 1076–1087 (2015). Ratcliffe, E. M. Intestinal microbiota influence the early
172. Jeffery, I. B. et al. An irritable bowel syndrome subtype postnatal development of the enteric nervous system. Author contributions
defined by species-specific alterations in faecal Neurogastroenterol. Motil. 26, 98–107 (2014). N.P. and M.M.W. researched data for the article, N.P. wrote
microbiota. Gut 61, 997–1006 (2012). 197. Luczynski, P. et al. Adult microbiota-deficient mice the article and N.P., M.M.W. and N.J.T. made substantial
173. Ikeda, M., Hamada, K., Sumitomo, N., Okamoto, H. have distinct dendritic morphological changes: contributions to discussion of content and reviewed or edited
& Sakakibara, B. Serum amyloid A, cytokines, differential effects in the amygdala and hippocampus. the manuscript before submission.
and corticosterone responses in germfree and Eur. J. Neurosci. 44, 2654–2666 (2016).
conventional mice after lipopolysaccharide injection. 198. Ford, A. C. et al. Effect of antidepressants and Competing interests statement
Biosci. Biotechnol. Biochem. 63, 1006–1010 (1999). psychological therapies, including hypnotherapy, The authors declare no competing interests.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY ADVANCE ONLINE PUBLICATION | 17


©
2
0
1
7
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