Вы находитесь на странице: 1из 15

REVIEW ARTICLE

published: 07 December 2011


doi: 10.3389/fphys.2011.00094

Braingutmicrobe communication in health and disease


Sue Grenham 1 , Gerard Clarke 1,2 , John F. Cryan 1,3 and Timothy G. Dinan 1,2 *
1
Laboratory of NeuroGastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
2
Department of Psychiatry, University College Cork, Cork, Ireland
3
Department of Anatomy, University College Cork, Cork, Ireland

Edited by: Bidirectional signalling between the gastrointestinal tract and the brain is regulated at
Stephen J. Pandol, University of
neural, hormonal, and immunological levels. This construct is known as the braingut axis
California at Los Angeles, USA
and is vital for maintaining homeostasis. Bacterial colonization of the intestine plays a major
Reviewed by:
Eileen F. Grady, University of role in the post-natal development and maturation of the immune and endocrine systems.
California at San Francisco, USA These processes are key factors underpinning central nervous system (CNS) signaling.
Govind K. Makharia, All India Institute Recent research advances have seen a tremendous improvement in our understanding
of Medical Sciences, India
of the scale, diversity, and importance of the gut microbiome. This has been reflected in
*Correspondence:
the form of a revised nomenclature to the more inclusive braingutenteric microbiota
Timothy G. Dinan, Department of
Psychiatry, GF Unit, Cork University axis and a sustained research effort to establish how communication along this axis con-
Hospital, Wilton, Cork, Ireland. tributes to both normal and pathological conditions. In this review, we will briefly discuss
e-mail: t.dinan@ucc.ie the critical components of this axis and the methodological challenges that have been pre-
sented in attempts to define what constitutes a normal microbiota and chart its temporal
development. Emphasis is placed on the new research narrative that confirms the critical
influence of the microbiota on mood and behavior. Mechanistic insights are provided with
examples of both neural and humoral routes through which these effects can be mediated.
The evidence supporting a role for the enteric flora in braingut axis disorders is explored
with the spotlight on the clinical relevance for irritable bowel syndrome, a stress-related
functional gastrointestinal disorder. We also critically evaluate the therapeutic opportuni-
ties arising from this research and consider in particular whether targeting the microbiome
might represent a valid strategy for the management of CNS disorders and ponder the
pitfalls inherent in such an approach. Despite the considerable challenges that lie ahead,
this is an exciting area of research and one that is destined to remain the center of focus
for some time to come.
Keywords: microbiota, central nervous system, enteric nervous system, irritable bowel syndrome, vagus nerve,
inflammation, probiotic, dysbiosis

INTRODUCTION communication along this axis contributes to both normal and


Scientific endeavor is increasingly characterized by a multidis- pathological conditions (Rhee et al., 2009).
ciplinary approach to the study of both health and disease. In this review, we will briefly discuss the critical compo-
Nowhere is this more evident than in the field of neurogas- nents of this axis and the methodological challenges that have
troenterology where the converging influence of experts across been presented in attempts to define what constitutes a normal
the diverse domains of gastroenterology, psychiatry, microbiol- microbiota and chart its temporal development. We examine the
ogy, pharmacology, immunology, and behavioral neuroscience, to approaches that have been taken to elucidate the impact of the
name but a few, have helped shape emerging biological themes. enteric microflora on this axis and vice-versa, with reference to
Chief among these is the concept of the braingut axis, a term the previously elucidated functions of the microbiota as well as an
which describes the complex bidirectional communication sys- evaluation of exciting new data suggesting a role for the microbiota
tem that exists between the central nervous system (CNS) and in the modulation of mood and behavior. Mechanistic insights
the gastrointestinal tract (GIT) and which is vital for maintain- are provided and the evidence supporting a role for the micro-
ing homeostasis (Cryan and OMahony, 2011). Spurred in part biota in disease states is discussed. The clinical implications are
by the discovery of Helicobacter pylori as a causative agent in critically evaluated, therapeutic opportunities arising from these
ulcer diseases but also by other innovative research in the gas- findings discussed and future perspectives are provided on this
trointestinal sciences (Pandol, 2010; Shanahan, 2010b), there is a rapidly expanding area of research.
growing appreciation of the critical role played by the commensal
microbiota, both in our general wellbeing and in the specific func- THE BRAINGUTENTERIC MICROBIOTA AXIS
tioning of the braingut axis. This has been reflected in the form The general scaffolding of the braingutenteric microbiota axis
of a revised nomenclature to the more inclusive braingutenteric includes the CNS, the neuroendocrine and neuroimmune sys-
microbiota axis and a sustained research effort to establish how tems, the sympathetic and parasympathetic arms of the autonomic

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 1


Grenham et al. Braingutmicrobe axis

nervous system (ANS), the enteric nervous system (ENS), and of As our knowledge of the previously unimagined scale of
course the intestinal microbiota. These components interact to microbial diversity within the gut has expanded, so too has an
form a complex reflex network with afferent fibers that project to understanding of its development and longitudinal variation.
integrative CNS structures and efferent projections to the smooth Colonization of the infant gut commences at birth when deliv-
muscle (OMahony et al., 2011). Put simply, through this bidi- ery exposes the infant to a complex microflora and its initial
rectional communication network, signals from the brain can microbiome has a maternal signature (see Figure 1; Mndar
influence the motor, sensory, and secretory modalities of the GIT and Mikelsaar, 1996; Mackie et al., 1999; Palmer et al., 2007).
and conversely, visceral messages from the GIT can influence brain The microbiome of unweaned infants is simple with high inter-
function (OMahony et al., 2011). This top-down and bottom-up individual variability (McCracken and Lorenz, 2001; Kurokawa
perspective of information flow as well as the detailed structural et al., 2007; Adlerberth and Wold, 2009). The numbers and diver-
integration and functioning of the various axis components has sity of strict anaerobes increase as a result of diet and environment,
been reviewed extensively elsewhere (Mayer, 2011). Less well stud- and after 1 year of age a complex adult-like microbiome is evident
ied but increasingly appreciated is the potential impact of the (Mackie et al., 1999; Kurokawa et al., 2007; Palmer et al., 2007).
enteric microbiota on the alliances within the construct (Rhee Of note is that a western diet is thought to be culpable for the
et al., 2009; Cryan and OMahony, 2011). development of a microbiota with a tendency toward an enhanced
proinflammatory motif (Greer and OKeefe, 2011; Wu and Hui,
MICROBIOTA COMPOSITION AND DEVELOPMENT 2011). Despite a significant interpersonal variation in the enteric
If size matters, then the numerical assessment of the microbiota microbiota, there seems to be a balance that confers health benefits
ensure that it wont be found wanting. The GIT is inhabited with and an alteration in beneficial bacteria can negatively influence the
1013 1014 microorganisms, a figure thought to be 10 times that wellbeing of the individual (Cryan and OMahony, 2011). Several
of the number of human cells in our bodies and 150 times as factors may alter the microbiome such as infection, disease, diet,
many genes as our genome (Gill et al., 2006; Qin et al., 2010). and antibiotics, but it tends to revert to the stable diversity estab-
The estimated species number varies greatly but it is generally lished in infancy once the threat of the initial distorting factor has
accepted that the adult microbiome consists of greater than 1000 subsided (Forsythe et al., 2010). Interestingly, it has been demon-
species (Qin et al., 2010) and more than 7000 strains (Ley et al., strated that the core microbiota of an aged individual is distinct
2006a). It is an environment dominated by bacteria, mainly strict from that of younger adults (Claesson et al., 2011) and that age
anaerobes, but also including viruses, protozoa, archae, and fungi related shifts in the composition of the intestinal microbiota are
(Gill et al., 2006; Xu et al., 2007). The microbiome is largely linked to adverse health effects in the elderly host (Woodmansey,
defined by two bacterial phylotypes, Bacteroidetes and Firmicutes 2007).
with Proteobacteria, Actinobacteria, Fusobacteria, and Verrucomi-
crobia phyla present in relatively low abundance (Eckburg et al.,
2005).
The surety with which these quantitative and qualitative
descriptions have been advanced belies the methodological dif-
ficulty encountered while trying to delineate and enumerate the
constituents of a normal microbiota. Culture based analyses,
the mainstay of traditional microbiological attempts to define
the enteric flora, are only adequate for the minority of the gut
microbiota that is amenable to cultivation (Eckburg et al., 2005;
Shanahan, 2010a). This problem has largely been circumvented
by the use of culture-independent techniques, a portfolio consist-
ing of sequencing based methods, genetic fingerprinting, fluores-
cently labeled oligonucleotide probes (FISH), quantitative PCR
as well as metagenomic approaches (Sekirov et al., 2010; Archie
and Theis, 2011). The realization that the secretory and meta-
bolic capability of the microbiome was likely as important as
phylotype composition has also led to the use of metabolomic
and metaproteomic approaches to improve our understanding
of what has been described as the forgotten organ (OHara and
Shanahan, 2006). Unfortunately, advances in culture methods
have not kept pace with the rise of these alternative technologies FIGURE 1 | Development of the microbiome in early life. Subsequent to
and a dual-pronged line of attack may be required to complete the sterile uterine environment, colonization begins at birth with facultative
the circle, a not inconsiderable logistical challenge (Clarke et al., bacteria (blue) colonizing the GIT immediately. The anaerobic bacteria
2009b). Moreover deficits remain to be redressed in relation to the colonize later (orange). By 1 year of age the microbiome has a stable
adult-like signature. Rodents follow a similar colonization pattern to humans
microbial content of the small intestine due to an over-reliance
and this forms the rationale for the use of germ free animals to study the
on the analysis of fecal microbial composition (Forsythe et al., impact of the microbiota.
2010).

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 2


Grenham et al. Braingutmicrobe axis

EVALUATING THE ROLE OF THE MICROBIOTA 1996; Stappenbeck et al., 2002). It is also clear from early stud-
Of the varied strategies employed to study the function of the ies that the microbiome is involved in maintenance of barrier
microbiota, perhaps the use of germ-free (GF) animals has offered function. Intestinal epithelial cell turnover is much slower in GF
the most revealing insights. Their use is based on the sterile animals than conventionally reared animals (Abrams et al., 1963).
uterine environment present during prenatal development (Adler- More recent studies substantiate these assertions with demon-
berth and Wold, 2009) with surgical delivery replacing the normal strations that commensal flora recognition by toll-like receptors
birthing process, thus eliminating the opportunity for post-natal (TLRs) is necessary to induce increased epithelial cell prolifer-
colonization of the GIT (see Figure 1). Subsequent comparison ation thus accelerating repair of the epithelial surface following
with their conventionally colonized counterparts allows infer- injury (Rakoff-Nahoum et al., 2004). Monoassociation of GF ani-
ences to be drawn regarding the morphological and physiological mals with Bacteroides thetaiotaomicron, a representative strain of
parameters that may be under the influence of the developing bacteria from the dominant microbial phyla, up-regulates sprr2a,
microbiota. However useful the complete absence of an intestinal a gene involved in desmosome maintenance and reinforcement of
microbiota may be in proof of principle studies, it is not reflective the intestinal barrier (Hooper et al., 2001). Furthermore, activa-
of real life scenarios. Other approaches which have more validity in tion of the TLR2 signaling pathway directly enhances intestinal
this regard involve inducement of a dysbiosis of the enteric flora, epithelial integrity through translocation of the tight junction
either through administration of antibiotics or deliberate infec- protein zonula occludens-1 (ZO-1; Cario et al., 2004). Paneth
tion in preclinical studies (Cryan and OMahony, 2011). Broad cells are secretory cells found in small clusters at the base of
spectrum antibiotics in particular are known to perturb the micro- crypts of Lieberkhn in the epithelium of the small intestine
biome by reducing biodiversity and delaying colonization and are and maintain intestinal homeostasis by directly sensing enteric
widely used as a method to intentionally alter the microbiome in bacteria at TLRs which trigger the expression of multiple anti-
a reproducible manner (Bennet et al., 2002; Donskey et al., 2003). microbial factors thus controlling intestinal barrier penetration
The use of cell lines, molecular approaches, and isografts have also by commensal and pathogenic bacteria (Vaishnava et al., 2008).
proved useful in evaluating the role of the microbiota (McCracken They are known to express and release a wide variety of anti-
and Lorenz, 2001). microbial peptides including -defensins and lysozyme C. but
require a complete microbiome to realize a full complement of
FUNCTIONAL RELEVANCE OF THE MICROBIOTA these peptides (Hooper et al., 2003; Cash et al., 2006; Vaishnava
GF studies have been pivotal in establishing the structural, protec- et al., 2008).
tive, and metabolic repertoire of functions that had been assigned GF studies also revealed that the microbiota is essential for the
to the microbiota even prior to the resurgence in interest in recent development of the gut associated lymphoid tissue (GALT) and
years (OHara and Shanahan, 2006). The morphological conse- indeed plays a vital part in shaping the immunological repertoire
quences of growing up germ-free were evidenced by the greatly of the GIT with IgA secretion and controlled inflammation being
enlarged cecum, reduced intestinal surface area, increased ente- regarded a consequence of bacterial colonization (Mayer, 2003;
rochromaffin cell area, smaller Peyers Patches and smaller villous Quigley, 2008). In comparison to conventionally housed animals,
thickness in these animals compared to conventional controls GF animals have decreased plasma cells and IgA, decreased expres-
(Wostmann and Bruckner-Kardoss, 1959; Gordon and Bruckner- sion of activation markers on intestinal macrophages, decreased
Kardoss, 1961; Abrams et al., 1963; Shanahan, 2002). It was perhaps MHCII on epithelial cells, decreased nitric oxide, and histamine
unsurprising, given these gross structural aberrations, that mul- levels in the small intestine (Gordon, 1959; Beaver and Wost-
tiple facets of normal GIT function would also be affected (see mann, 1962; Glaister, 1973; Moreau et al., 1978; Matsumoto
Figure 2). et al., 1992; Mikkelsen et al., 2004; Sobko et al., 2004; Smith
Thus it is known that the microbiota is essential for normal et al., 2007). Peyers patch follicles are reduced in number and
GIT motility, with deficits due in part to perturbations in peristal- size and the mesenteric lymph nodes are smaller, less cellu-
sis on the back of impaired smooth muscle layer function (Berg, lar, and do not have germinal centers in GF animals (Gordon,
1959; Glaister, 1973). However, reconstitution of GF mice with an
intestinal microflora is sufficient to restore the mucosal immune
system (Umesaki et al., 1995). Moreover, ligands from commen-
sal bacteria such as polysaccharide A, lipopolysaccharide (LPS),
and lipoteichoic acid (LTA) influence the normal development
and function of the mucosal immune system (Rakoff-Nahoum
et al., 2004; Mazmanian et al., 2005). The basic mechanism of
the mucosal immune system is innate immunity and its char-
acteristic ability to distinguish potentially pathogenic microbes
from harmless antigens is achieved through pattern recognition
receptors (PRR). TLRs are present on cells of the innate immune
system and recognize characteristic molecules called pathogen
FIGURE 2 | Function of the intestinal microbiome. Commensal bacteria associated molecular patterns (PAMPS; Akira and Hemmi, 2003).
exert a miscellany of protective, structural, and metabolic effects on the
Pathogen recognition by a particular TLR results in a cascade of
intestinal mucosa.
events starting with the activation of the NF-B signaling system

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 3


Grenham et al. Braingutmicrobe axis

and resulting in increased cytokine production and T cell acti- compared to the specific pathogen free (SPF) controls. The stress
vation (Ulevitch, 1999). However in the absence of the resident response in the GF mice was partially reversed by recolonization
enteric flora, key members of the TLR family have low or absent with fecal matter from SPF animals and fully reversed by monoas-
expression profiles in the GIT, thus compromising appropriate sociation with B. infantis in a time dependant manner (Sudo
immune responses to pathogenic threats (OHara and Shanahan, et al., 2004). This study clearly demonstrated that the microbial
2006). content of the GIT is critical to the development of an appro-
The microbiota also has a critical role in supporting normal priate stress response later in life and also that there is a critical
digestion and host metabolism. There are two main mechanisms window in early life where colonization must occur to ensure
by which it can maximize nutrient availability, either by the release normal development of the HPA axis. Sudo et al. (2004) also
of calories from otherwise unavailable oligosaccharides or by mod- reported a decrease in brain derived neurotrophic factor (BDNF),
ulating absorption (Sekirov et al., 2010). A significant energy a key neurotrophin involved in neuronal growth and survival,
source for humans is the bacterial metabolism of dietary fiber to and expression of the NMDA receptor subunit 2a (NR2a) in
short-chain fatty acids (SCFAs; Macfarlane and Macfarlane, 2003). the cortex and hippocampus of GF animals compared to SPF
SCFAs can modulate the host energy balance through Gpr41, a G controls.
protein coupled receptor that binds SCFA, and is dependent upon The encouraging findings from this study prompted further
the gut microbiome (Samuel et al., 2008). It is thought that inter- research in this area, especially with a view to establishing a
action between SCFAs produced by the gut bacteria, and Gpr41 behavioral phenotype and neurochemical profile that might be
increases circulating levels of PYY, an enteroendocrine hormone associated with the gut flora. To date, the most consistent data
that reduces gut motility and thus increases absorption of SCFAs has been in relation to indices of anxiety. Neufeld et al. (2011),
(Samuel et al., 2008). GF animals require a higher caloric intake despite reporting an unexplained increase in BDNF mRNA that
to maintain the same body weight as conventional animals and was contrary to the protein decreases observed in the earlier study,
are prone to vitamin deficiencies, requiring dietary supplementa- reported a less anxious phenotype for germ free animals in the ele-
tion with vitamins K and B (Sumi et al., 1977; Wostmann, 1981; vated plus maze (EPM). This behavioral phenotype was replicated
Wostmann et al., 1983). in another study with GF animals in both the EPM and the light
Prevention of colonization by pathogens is achieved in large dark box and associated with an altered gene expression profile in
part through the resident microbiota by competing for nutri- relevant brain regions (Heijtz et al., 2011). Probiotic administra-
ents and receptors and production of anti-microbial compounds tion studies also weigh in on the side of a role for the microbiota
(Sekirov et al., 2010). Lactobacillus and Bifidobacterium are gram in anxiety-like behaviors with, for example, administration of L.
positive bacteria and form two important genera in the micro- helveticus R0052 and B. longum R0175 taken in combination dis-
biome. These bacteria were shown to inhibit listerial infections playing anxiolytic-like activity in rats (Messaoudi et al., 2011).
in vitro. It is thought that Lactobacilli inhibited infection through Moreover, a recent study reported that chronic treatment with
a combination of acid production and secretion of an uniden- the probiotic L. rhamnosus (JB-1) over 28 days produced animals
tified protein while Bifidobacterium inhibition was attributed to with lower levels of stress-induced corticosterone and reduced
an extracellular proteinaceous secreted compound (Corr et al., depressive behaviors in the forced swim test in addition to a less
2007a). In another study a Lactobacillus salivarius strain was shown anxious phenotype in the EPM. The L. rhamnosus (JB-1) treated
to produce a bacteriocin in vivo that could significantly protect animals also showed alterations of GABAB1b mRNA in the brain
mice against infection with the pathogen Listeria monocytogenes with increased expression in cortical regions and decreased expres-
(Corr et al., 2007b). sion in the hippocampus, amygdala, and locus coeruleus as well
as reduced GABAA2 mRNA expression in the prefrontal cortex
MICROBIOTA AND THE CNS and amygdala and increased GABAA2 in the hippocampus. Inter-
It is clear then from the gamut of functions under the influence of estingly the authors demonstrated that vagotomized mice did not
the microbiota regulation of the mucosal immune system, GIT display the neurochemical and behavioral effects of this bacterium,
motility, and epithelial barrier function, support for digestion/host thus implicating the vagus nerve in the direct communication
metabolism and prevention of colonization by pathogens that between the bacteria and the brain (Bravo et al., 2011). A role
critical components of the braingut axis other than the GIT itself for the gut microbiota in pain perception has also been proposed
might also be subject to manipulation by the microbiome. More (Forsythe et al., 2010), with, for example, one study demonstrating
specifically, the potential influence of the commensal bacteria on that specific Lactobacillus strains could induce the expression of -
CNS signaling is currently, after a slow start, a burgeoning area of opioid and cannabinoid receptors in intestinal epithelial cells and
research. mimic the effects of morphine in promoting analgesia (Rousseaux
The core neuroendocrine pathway in man is the hypothalamic et al., 2007).
pituitaryadrenal (HPA) axis and activation of this axis takes place A strategy employing antibiotic-induced dysbiosis of the
in response to a variety of physical and psychological stressors microbiome, in this case a cocktail consisting of neomycin, baci-
(Dinan et al., 2006). After much initial speculation, an elegant tracin, and the antifungal agent pimaricin, resulted in mice which
study by Sudo et al. (2004) provided some insight into the role of displayed less anxiety-like behaviors in the both the step down
the intestinal microbiota in the development of the HPA axis. In box and the light/dark box test. Interestingly the authors also
GF mice a mild restraint stress induced an exaggerated release reported altered protein levels of BDNF in the amygdala and
of corticosterone and adrenocorticotrophin hormone (ACTH) hippocampus and that discontinuation of the antibiotic cocktail

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 4


Grenham et al. Braingutmicrobe axis

restored the normal behavioral profile of the animals (Bercik et al.,


2011). Similarly perturbation of the microbiota by means of an
infectious agent like Citrobacter rodentium has been shown to
increase anxiety-like behavior in mice 78 h post infection as
measured in the hole board open field apparatus (Lyte et al.,
2006) and to produce stress-induced memory dysfunction 10 and
30 days post infection (Gareau et al., 2011). Moreover, memory
dysfunction was prevented by daily administration of a probi-
otic cocktail and when GF mice were infected they developed
memory dysfunction regardless of whether they were stressed
or not.

INFLUENCE OF THE BRAIN ON THE MICROBIOME


Although the bulk of research to date has focused on the impact
of the microbiota on CNS function, there is also research to sug-
gest that the brain can alter the microbiome. Signaling molecules
released into the gut lumen from cells in the lamina propria
that are under the control of the CNS can result in changes in
gastrointestinal motility and secretion as well as intestinal perme-
ability, thus altering the GIT environment in which the bacteria
reside (Rhee et al., 2009). Stress also induces permeability of the
gut allowing bacteria and bacterial antigens to cross the epithe-
lial barrier and this can activate a mucosal immune response
which in turn alters the composition of the microbiome (Kili-
aan et al., 1998). Acute stress was shown to cause an increase
in colonic paracellular permeability which involved mast cells FIGURE 3 | Proposed mechanisms of action. There are a variety of
and overproduction of IFN- with decreased expression of ZO-2 proposed mechanisms, including both humoral and neural routes, through
and occludin mRNA (Demaude et al., 2006). The psychological which the microbiota can modulate signaling along the braingut axis. For
components of social stress was shown to facilitate the translo- example, recent studies suggest a role for both the vagus nerve and
modulation of systemic tryptophan levels in relaying the influence of both
cation of indigenous bacteria into the host (Bailey et al., 2006).
resident and exogenous microflora along this bidirectional communication
Other studies have shown that stress hormones promoted the axis.
growth of non-pathogenic isolates of Escherichia coli as well as
the pathogenic E. coli 0157:H7 strain via interactions with host
catecholamines such as adrenaline and noradrenaline (Freestone study which conclusively implicated the vagus nerve in the direct
et al., 2002, 2003). Different psychological stressors are known communication observed between the bacteria and the brain (see
to alter the composition of the microbiome by modulating the earlier sections for detailed description; Bravo et al., 2011). Fur-
composition of total biomass in infants (Rhee et al., 2009). Pre- ther studies are required to clarify the precise route though which
natal stressors have been shown to alter the microbiome in rhesus the administered probiotic interacts with the vagus nerve but in
monkeys by reducing the overall numbers of Bifidobacteria and any case this study confirmed a mechanism many had previously
Lactobacilli (Bailey et al., 2004). Maternal separation, an early suggested as a likely contender (Cryan and OMahony, 2011).
life stressor, caused a significant decrease in fecal Lactobacilli on Other potential mechanisms of action include neurotransmitter
day 3 post separation, which returned to baseline by day 7 as modulation. B. infantis 35624, for example, has been shown to
assessed by enumeration of total and gram-negative aerobic and induce an elevation in plasma tryptophan levels, a precursor to
facultative anaerobic bacterial species (Bailey and Coe, 1999). serotonin (5-HT) which is a key neurotransmitter within the
However, early life stress can also have long term effects on the braingut axis, in Sprague-Dawley rats (Desbonnet et al., 2008).
microbiome. Analysis of the 16S rRNA diversity in adult rats Since CNS tryptophan concentrations are largely dependant on
exposed to maternal separation for 3 h per day from post-natal peripheral availability and the enzymatic machinery responsible
days 212 revealed a significantly altered fecal microbiome when for the production of 5-HT is not saturated at normal tryptophan
compared to the non-separated control animals (OMahony et al., concentrations (Ruddick et al., 2006), the implication here is that
2009). the microbiota might play some role in the regulation of CNS as
well as ENS 5-HT synthesis. This effect is potentially mediated
MECHANISMS OF ACTION by the effect of the microbiota on the expression of indoleamine-
A number of mechanisms have been proposed through which the 2,3-dioxygenase, a key enzyme in the physiologically dominant
intestinal communal microflora might influence ENS and CNS kynurenine pathway of tryptophan degradation (Forsythe et al.,
signaling, including both neural and humoral routes as well as 2010). Of course multiple mechanisms are possible and indeed
direct and indirect modes of action (see Figure 3; Forsythe et al., likely, given the strain specific effects that have been observed in
2010). Perhaps the best evidence to date comes from a novel recent many probiotic studies to date (Quigley, 2008).

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 5


Grenham et al. Braingutmicrobe axis

MICROBIOTA IN DISEASE controls (Ley et al., 2005). In humans, when the fecal microbiota
Since the microbiota is involved in maintaining homeostasis and of obese individuals was compared to lean controls there were
wellbeing, the proposition that it may also be involved both in gas- fewer Bacteroides and more Firmicutes in the obese group. How-
trointestinal and systemic illnesses is a logical one (see Figure 4). ever, over time on a calorie restricted diet the relative abundance of
As might be expected given the importance of the microbiota in Bacteroidetes increased and the abundance of Firmicutes decreased
supporting host digestion and metabolism, obesity has been con- (Ley et al., 2006b). While these studies do offer some support
sidered as an illness with a potential microbial basis. It has been for a purturbations of the resident flora in the accumulation
defined as a medical condition in which excess body fat has accu- of excess fat, microbial influences should not be considered in
mulated to produce adverse effects on health, leading to increased isolation: obesity is a multifactorial condition that also involves
health problems and potentially a reduced life expectancy (Haslam strong genetic factors, hypothalamic dysfunction, and an increase
and James, 2005). The incidence of obesity is increasing in both in the consumption of energy-dense food (Schellekens et al.,
developed and developing countries and has become a worldwide 2010).
burden upon health care systems (Bloom et al., 2008). Some of Less intuitive is a potential role for the microbiota in the devel-
the most convincing evidence comes from studies in GF ani- opment of autism, a developmental disorder that appears in the
mals which have shown them to be naturally leaner than their first 3 years of life and affects the brains normal development of
SPF counterparts and colonization of adult GF mice with a nor- social and communication skills (American Psychiatric Associa-
mal microbiota from the cecum of conventionally raised animals tion, 2000). Interestingly in a study of 58 autism patients >90%
produces a 60% increase in body fat content and insulin resis- had gastrointestinal problems compared to none in the control
tance within 14 days despite reduced food intake (Bckhed et al., group (Parracho et al., 2005). Although the underlying etiology of
2004). GF mice are resistant to diet induced obesity but when col- autism is not well understood, late onset autism is hypothesized
onized with fecal flora from SPF mice they gained weight due to be caused by extensive use of antibiotics with an association
to altered fatty acid metabolism (Bckhed et al., 2007). Other with Trimethoprim/sulfamethoxazole antibiotic treatment (Bolte,
studies have shown that the ob/ob mouse, a strain of mouse 1998; Finegold et al., 2002). There is evidence to support alter-
genetically predisposed to obesity, had a 50% decrease in the ations of the fecal microbiota in patients with autism, with an
abundance of Bacteroidetes and a proportional increase in Fir- increase in several subtypes of Clostridium (Song et al., 2004;
micutes when compared to the lean wild type and heterozygotic Finegold et al., 2010) and in the majority of cases treatment with
vancomycin, an antibiotic that targets gram positive anaerobes and
is minimally absorbed by the GIT, can improve symptoms (Sandler
et al., 2000). It is further hypothesized that Clostridium spores are
responsible for relapse after completion of the antibiotic treatment
and that a treatment targeting these spores alleviates symptoms for
longer periods (Finegold, 2008).
Within the realm of gastrointestinal disorders, inflammatory
bowel disease (IBD), which includes the two distinct disease pat-
terns of ulcerative colitis (UC) and Crohns disease (CD; Lichten-
stein, 2000; Blumberg and Strober, 2001; Podolsky, 2002; MacDon-
ald and Monteleone, 2005), has attracted attention as a disorder
with an aberrant GIT microbial signature (Shanahan, 2004). Sev-
eral studies have shown altered gut microbiota in patients with
IBD although it is not clear whether these changes are responsible
for causing disease or are the result of inflammatory responses and
extensive tissue changes in the GIT (Macfarlane et al., 2009). This
was not clarified by a study which showed that the microbiome was
also significantly altered in mucosal biopsies taken from inflamed
compared to healthy sites (Walker et al., 2011). The changes in the
microbiota are characterized by increases in Proteobacteria and a
decline in Firmicutes and Bacteroidetes (Frank et al., 2007; Walker
et al., 2011). This altered microbiota is also evident in animal mod-
els of inflammation relevant to IBD with a dramatic increase in
FIGURE 4 | Braingutmicrobe communication in health and disease. A
the Proteobacteria classes of bacteria evident (Lupp et al., 2007).
stable gut microbiota is essential for normal gut physiology and contributes
to appropriate signaling along the braingut axis and to the healthy status of
Interestingly although a Bacteroides species has been shown to col-
the individual as shown on the left hand side of the diagram. Conversely, as onize both genetically IBD susceptible and non-susceptible mice
shown on the right hand side of the diagram, intestinal dysbiosis can to the same extent, it only engendered disease in the susceptible
adversely influence gut physiology leading to inappropriate braingut axis animals (Bloom et al., 2011). Furthermore, in terms of exoge-
signaling and associated consequences for CNS functions and disease
nous microbial threats, the frequency of Clostridium difficile has
states. Stress at the level of the CNS can also impact on gut function and
lead to perturbations of the microbiota. been shown to be higher in IBD and may trigger relapse where
the disease is established but in remission (Clayton et al., 2009).

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 6


Grenham et al. Braingutmicrobe axis

Although the pathogenesis of IBD is still not completely under- increased numbers of activated mast cells in colonic biopsies from
stood, it is well recognized that psychological stress, a factor which IBS patients and a direct correlation between proximity of the
can perturb the microbiota, exacerbates the condition (Mawdsley mast cells to neurons and pain severity (Barbara et al., 2004).
and Rampton, 2006; Reber, 2011). However it should be recog- Cenac et al. (2007) demonstrated increased proteolytic activity in
nized that IBD has a complex and multifaceted etiology, involving colonic washes from IBS compared with control patients while the
environmental and genetic factors which cause dysregulation of analysis of TLR expression in colonic biopsies has demonstrated
the mucosal immune system (Andus and Gross, 2000; MacDon- increased expression of TLR4 and TLR5 and decreased levels of
ald and Monteleone, 2005). Thus, while the microbiota might TLR7 and TLR8 in IBS patients (Brint et al., 2011).
not be the sole driving force in IBD, most consider that host Others have investigated the involvement of the systemic
flora interactions underpin the disorder, especially in genetically immune system on the basis that it might reflect mucosal
susceptible individuals (Shanahan, 2005; Melgar and Shanahan, disturbances. In a study of 78 IBS patients OMahony et al.
2010). (2005) demonstrated an abnormal IL-10/IL-12 ratio in stimulated
The best evidence to date for the involvement of the microbiota peripheral blood mononuclear cells (PBMCs), indicating a proin-
in disease states comes from irritable bowel syndrome (IBS), a flammatory, Th-1 state. Liebregts et al. (2007) showed an enhanced
prototypical stress-related braingut axis disorder. IBS is the most proinflammatory cytokine release (TNF-, IL-1, IL-6) in LPS
common functional gastrointestinal disorder (FGID) encountered stimulated PBMCs from IBS patients. In another study, baseline
in clinical settings (Camilleri, 2001), affecting an estimated 10 plasma proinflammatory cytokines IL-6, IL-6R, and IL-8 were ele-
15% of the general population in Western Europe and North vated in IBS and accompanied by overactivation of the HPA axis
America (Quigley, 2011). It is characterized by the presence of (Dinan et al., 2006). Indeed elevations in systemic proinflamma-
abdominal pain or discomfort, an alteration in bowel habit, and tory mediators have repeatedly being demonstrated in both clinical
the absence of reliable biomarkers (Thompson et al., 1999; Dross- datasets and reports from animal models of the disorder (Dinan
man et al., 2002; Clarke et al., 2009b). A number of strands of et al., 2008; Clarke et al., 2009a, 2010; OMahony et al., 2009;
evidence support a role for the microbiota in the pathophysi- Scully et al., 2010). What is not clear is whether they truly rep-
ology of IBS and chief among these is the supporting data for resent a disturbance at mucosal sites and this remains a subject
post-infectious IBS (PI-IBS), a term which describes the develop- in need of further investigation. Interestingly it has recently been
ment of IBS following an episode of bacteriologically confirmed reported that IBS patients have a distinct pattern of exaggerated
gastroenteritis (Quigley, 2009; Sarna, 2011). This phenomenon is peripheral TLR activity as indicated by measurements of cytokine
now well documented (Gwee et al., 1999, 2003; Rodrguez and production following whole blood stimulations (McKernan et al.,
Ruigmez, 1999; Marshall et al., 2006, 2007) and prospective stud- 2011).
ies showing that the risk of developing PI-IBS is in the order of The qualitative alterations and temporal instability that have
336% following an enteric infection with the precise incidence been reported in the enteric flora in IBS cohorts, summarized in
depending on the infecting organism (Spiller and Garsed, 2009). Table 1, is taken as further evidence of an involvement in disease
The highest reported incidence of PI-IBS was associated with the pathophysiology and is backed up by data from animal models
Walkerton outbreak with 36% of individuals showing symptoms of the disorder (OMahony et al., 2009; Quigley, 2009). While it is
of IBS 2 years after infection with both Campylobacter jejuni and unclear at present is whether these changes in the flora are primary
E. coli 0157:H7 (Marshall et al., 2006). A mechanistic perspective or secondary and although they have proven difficult to repro-
has been provided by a number of studies which found a persistent duce in many cases, there are certainly routes through which they
elevation in rectal mucosal enteroendocrine cells, T-lymphocytes could impact on symptom pathogenesis (Quigley, 2009). Quanti-
and gut permeability following the infectious insult in subjects tative alterations in the flora, which have centered around claims
who went on to develop IBS (Spiller et al., 2000; Dunlop et al., of small intestinal bacterial overgrowth (SIBO) and have in some
2003). These studies are regarded as important indicators of a link cases formed the proposed basis for the efficacy of antibiotics such
between alterations in the microbiota and mucosal inflammation as rifaximin in IBS, are more controversial with critics question-
in IBS (Quigley, 2009). ing both the specificity and validity of the measurements (Quigley,
On the basis that immunological changes in IBS might result 2007). This is also contrary to the view that antibiotics might be a
from exposure to exogenous bacterial challenge, studies sup- risk factor for the development of IBS (Collins et al., 2009).
porting the low grade inflammation that has so frequently been
reported in IBS are also taken as evidence of a role for the micro- THERAPEUTIC OPPORTUNITIES
bial perturbations in the disorder (Clarke et al., 2009b; Quigley, Although modulation of the microbiota has primarily been a
2009). Persistent low grade inflammation is a characteristic of PI- source of evidence for the functional importance of the commen-
IBS (Gwee et al., 1999) and these patients exhibit greater IL-1 sal load in health, the swell of studies implicating our resident
mRNA expression, both during and after the infection, com- microflora in disease states such as IBS means that this could also
pared with individuals who do not develop PI-IBS (Gwee et al., be a strategy with therapeutic potential. There is mounting evi-
2003). IBS patients with normal histology had increased intraep- dence to support the use of antibiotics, prebiotic substances, and
ithelial lymphocytes and CD3+ and CD25+ cells in the lamina probiotics with the latter option in particular enjoying some suc-
propria (Chadwick et al., 2002). Increased CD25+ cells in IBS cess as a treatment option in IBS (Quigley, 2008; Rhee et al., 2009).
suggests an antigen challenge and these cells are preventing a Thus, although the effects appear to be strain specific, cumula-
more florid inflammatory response (Collins, 2002). There are tive data from the clinical trials to date suggest a role for certain

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 7


Grenham et al. Braingutmicrobe axis

Table 1 | Studies which have shown altered fecal and mucosal microbiome in disease states.

Group Method, microbiota analyzed Diagnostic criteria and subjects Finding Reference

IBS Q-PCR, fecal microbiome Rome II, n = 27 Lactobacillus spp. in IBS-D subjects Malinen
BS-D (n = 12), IBS-C (n = 9), Veillonella spp. in IBS-C et al. (2005)
IBS-A (n = 6)
Culture/DGGE, fecal Rome II, n = 26 Increased number of aerobes in IBS patients Mtt et al.
microbiome BS-D (n = 12), IBS-C (n = 9), Temporal instability in IBS patients revealed by DGGE (2005)
IBS-A (n = 5)
Q-PCR, Phylogenic Rome II, n = 62 Ratio of the Firmicutes to Bacteroidetes in IBS Rajilic-
Microarray, fecal microbiome IBS-D (n = 25), IBS-C (n = 18), in numbers of Dorea, Ruminococcus, and Stojanovic
IBS-A (n = 19) Clostridium spp. in IBS et al., 2011)
Bacteroidetes in IBS
Bifidobacterium and Faecalibacterium spp.
Average number of methanogens in IBS
Fractionation/16S rRNA gene Rome II, n = 24 Significant differences in Coprococcus, Collinsella, and Kassinen
cloning and sequencing, IBS-D (n = 10), IBS-C (n = 8), Coprobacillus Phyla in the IBS group compared to et al. (2007)
Q-PCR, fecal microbiome IBS-A (n = 6) controls
DGGE, fecal microbiome Rome II, n = 47 Significant difference between IBS and healthy Codling
controls et al. (2010)
No sub-typing Significantly more variation in microbiota of healthy
volunteers than that of IBS patients
FISH, fecal and duodenal Rome II, n = 41 Bifidobacteria in IBS subjects compared to healthy Kerckhoffs
microbiome IBS-D (n = 14) IBS-C (n = 11) controls et al. (2009)
IBS-A (n = 16)
Q-PCR, fecal microbiome Rome II and III, n = 26 Veillonella and Lactobacillus in IBS Tana et al.
IBS-D (n = 8) IBS-C (n = 11) IBS-A (2010)
(n = 7)
IBD FISH adapted to flow Active CD (n = 13) Clostridium coccoides was reduced in UC Sokol et al.
cytometry, fecal microbiota Active UC (n = 13) C. leptum group was reduced in CD (2006)
IC (n = 5), HS (n = 13). Bacteroides group was more abundant in IC
16S rRNA DGGE analysis, Active CD (n = 5) Temporal stability of dominant species for all Crohns Scanlan
fecal microbiota Inactive CD (n = 11), HS (n = 18) disease patients et al. (2006)
Bifidobacterium spp. were similar in all samples
Clostridiales and Bacteroidales communities are
altered in Crohns disease
DGGE, Q-PCR, fecal CD (n = 68) CD vs. unaffected relatives Joossens
microbiota Unaffected relatives (n = 84), HS Dialister invisus, an uncharacterized species of et al. (2011)
(n = 55) Clostridium cluster XIVa, Faecalibacterium prausnitzii
and Bifidobacterium adolescentis
Ruminococcus gnavus
Unaffected relatives vs. HS
Collinsella aerofaciens and member of the
Escherichia coliShigella group
Ruminococcus torques
T-RFLP analysis16S rRNA Monozygotic twin pairs that Predominantly ileal CD vs. co-twins and CD localized Willing et al.
gene, Q-PCR, ileal and rectal were discordant (n = 6) or in the colon (2009)
biopsies concordant (n = 4) for CD, HS Faecalibacterium prausnitzii
(n = 6) Escherichia coli
16S rRNA gene sequencing, Inflamed and non-inflamed Mucosal microbial diversity in IBD Walker et al.
mucosal biopsies intestinal tissue from 6 CD Firmicutes in IBD samples and Bacteroidetes (2011)
(n = 12), 6 UC (n = 12), HS (n = 5) Enterobacteriaceae in CD only significant
differences in microbial community structure between
inflamed and non-inflamed mucosal sites
rRNA sequence analysis and UC (n = 61), CD (n = 68) Bacteroidetes and Lachnospiraceae in IBD Frank et al.
Q-PCR HS (n = 61) Actinobacteria and Proteobacteria in IBD (2007)

(Continued)

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 8


Grenham et al. Braingutmicrobe axis

Table 1 | Continued

Group Method, microbiota analyzed Diagnostic criteria and subjects Finding Reference

Obesity Pyrosequencing, cecal Ob/Ob mice Firmicutes Turnbaugh


contents Firmicute to Bacteroides ratio et al. (2006)
16S rRNA sequencing, cecal Ob/Ob mice Bacteroidetes ob/ob animals Ley et al.
contents Firmicutes (2005)
16S rRNA sequencing Human Bacteroides in obesity Ley et al.
Bacteroides during calorie restriction (2006b)
Q-PCR, MALDI-TOF spectral Human Bifidobacterium animalis and Methanobrevibacter Million et al.
analysis, fecal microbiota smithii were associated with normal bodyweight (2011)
Lactobacillus reuteri was associated with obesity
Autism Q-PCR, fecal microbiota Autistic (n = 15), HS (n = 8) Clostridium bolteae and Clostridium clusters I and Song et al.
XI in autistic children (2004)
Clostridium cluster XIVab in autistic children
16S rRNA gene sequencing Autistic (n = 13), HS (n = 8) Fecal samples Finegold
and culture, fecal microbiota Children with autism had nine species of Clostridium et al. (2002)
Gastric and duodenal not found in controls
sampling Control children had three species of Clostridium not
found in autistic children Gastric and duodenal
Specimens
No non-spore-forming anaerobes and microaerophilic
bacteria from controls significant numbers of
non-spore-forming anaerobes and microaerophilic
bacteria in children with autism
Pyrosequencing, fecal Autistic (n = 33) Increased diversity and richness in the autistic Finegold
microbiota Non-affected siblings (n = 7), HS gastrointestinal microbiome et al. (2010)
(n = 8) Bacteroidetes in the severely autistic group
Firmicutes in the control group
Desulfovibrio species and Bacteroides vulgatus
autistic children

IBS-D, diarrhea predominant IBS; IBS-C, constipation predominant IBS; IBS-A, alternating IBS; UC, ulcerative colitis; CD, Crohns disease; IC, infectious colitis; HS,
healthy subjects; DGGE, denaturing gradient gel electrophoresis; Q-PCR, quantitative PCR; FISH, fluorescent in situ hybridization; T-RFLP, terminal restriction length
polymorphism.

probiotic therapies in the alleviation of key symptoms in IBS braingut axis dysfunction (Desbonnet et al., 2010; OMahony
(Abdominal pain/discomfort, bloating/distension, alterations in et al., 2011). Of further interest is that a preclinical study using the
defecatory function; Moayyedi et al., 2008; Brenner et al., 2009; same probiotic found that CRD-induced visceral pain behaviors
Whorwell, 2009). were significantly reduced in the viscerally hypersensitive Wistar-
Recent research supporting a role for the microbiota in main- Kyoto rat strain (McKernan et al., 2010). L. paracasei NCC2461
taining normal brain function offers the intriguing possibility administration has also shown efficacy in reducing visceral hyper-
that the therapeutic targeting of the gut microbiome might be sensitivity in a mouse preclinical model of IBS (Verdu et al.,
a viable strategy in the treatment of CNS disorders (Forsythe 2006).
et al., 2010). Some validation for this approach can be seen in the Few studies have made an attempt to unravel the mystery
results of a recent study which demonstrated beneficial psycholog- surrounding the mechanism of action of probiotics. However a
ical effects in healthy human volunteers following administration number of theories exist, many of which relate to the previously
of a combination of L. helveticus R0052 and B. longum R0175 elucidated role of the microbiota in health. The possibility that
(Messaoudi et al., 2011). However this field of research is still in the administered probiotic might restore the distorted bacterial
its infancy with few studies exploring the concept of microbial flora to one resembling that of healthy individuals is partially
targeting of the GIT under pathological conditions of the CNS. supported by a study demonstrating exactly such a phenomenon
Nevertheless, studies that have employed this strategy have gener- with a multispecies probiotic preparation previously shown to
ated encouraging results. B. infantis 35624 treatment, for example, be effective in the treatment of IBS (Kajander et al., 2005; Lyra
was shown to normalize immune responses, reverse behavioral et al., 2010). Similarly alterations in the fecal metabolome follow-
deficits in the forced swim test, and restore basal noradrena- ing administration of an alternative probiotic preparation were
line concentrations in the brainstem of adult rats subjected to linked to its previously demonstrated efficacy (Hong et al., 2009,
the early life stress of maternal separation, an animal model of 2011). However experts in the field consider such a mechanism,

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 9


Grenham et al. Braingutmicrobe axis

along with the assumption of competition with and exclusion some way from where this will be a serious issue for the appli-
of pathogens, to be overly simplistic and undermined by an cation of microbiome manipulation to the treatment of specific
incomplete knowledge of what constitutes a normal microbiome CNS disorders but it is already a major impediment facing those
(Quigley, 2009). who are currently seeking to use the approach for gastrointestinal
The notion that efficacious probiotic strains might exert their afflictions.
beneficial effects through modulation of host immunity is gain-
ing traction, especially in the light of the expanding repertoire of CONCLUSION AND FUTURE PERSPECTIVES
studies supporting a low grade inflammation in IBS (Clarke et al., The microbiome has assumed its rightful position as a critical
2009b). Of the strains evaluated, B. Infantis 35624 has been shown component of the braingut axis and a key factor in both health
to normalize abnormal IL-10/IL-12 ratios in addition to showing and disease, a fact reflected in current research efforts. Previ-
efficacy in treatment of the disorder (OMahony et al., 2005). That ously determined functions of the microbiota have been bolstered
certain probiotics might work by enhancing barrier function and with exciting new studies highlighting its potential involvement
motility has also been considered and is supported primarily by in a range of disorders as well as its impact on behavior and
data from Ussing chamber studies. B. breve C50 and its soluble mood at the level of the CNS. Recent findings raise the possi-
factors have been shown to alleviate chloride secretion in human bility that therapeutic targeting of the microbiome might be an
intestinal epithelial cells (Heuvelin et al., 2009). Bioactive peptide effective treatment strategy for specific disorders of the CNS. This
factors secreted by probiotics have been shown to enhance epithe- needs to be tempered by the realization that we are still some way
lial cell barrier function both in vitro and in vivo (Ewaschuk et al., from being able to completely differentiate a normal microbiota
2008). Pretreatment with a probiotic mixture containing L. helveti- from that present in disease or recognizing how its individual
cus and L. rhamnosus was shown to prevent bacterial translocation components and collective signature are critical for health. Fur-
and improve intestinal barrier function in rats following chronic thermore although recent mechanistic insights have provided a
psychological stress (Zareie et al., 2006). Similarly, L. Plantarum solid platform for future studies, an advanced understanding of
299V was shown to inhibit Escherichia coli-induced intestinal per- the interactions that occur is still lacking and warrants further
meability (Mangell et al., 2002). The involvement of the vagus research. Whether the approach of microbiome manipulation will
nerve is also considered likely in the light of recent studies (Bravo evolve into a therapy in its own right or become an adjuvant to
et al., 2011; Cryan and OMahony, 2011). In reality the mecha- conventional therapy is also still unclear (Aragon et al., 2010).
nism of action is likely to be multifactorial with particular strains While this will involve clarification on the specific assessment cri-
exerting their influence either though one or more of the modes teria for successful health claims by agencies like FDA and EFSA,
outlined above or via a yet unidentified means. it also remains to be seen whether probiotic manufacturers will
Despite the demonstrated efficacy of microbiota manipulation be willing to subject their products to assessment by the rigor-
in clinical disorders of the GIT, the potential utility of the strat- ous clinical trials that would be necessary for specified use of their
egy in the treatment of CNS disorders as gleaned from preclinical products in the treatment of disease states. In this context, support-
data and an improved understanding of the mechanisms involved, ing gut health through microbiota supplementation with a view
a number of caveats still exist surround this approach generally. toward positively influencing mental status represents a putative
One such limitation is the mixed results that have been obtained preventative strategy worth following (Bischoff, 2010). In addi-
in organic disorders of the GIT such as IBD where controlled tri- tion to consideration of the microbiome as a therapeutic target,
als in CD have generally been disappointing as against the more we also need to direct more efforts toward mining its metabo-
favorable results in UC (Shanahan, 2010b). There is also a paucity lites for putative drugs, a strategy that has already paid some
of data pertaining to the long term effects of microbiome manip- dividends (Shanahan, 2010a). It is unmistakable that braingut
ulation although it is encouraging to note that a trial with a mul- microbiome communication in health and disease will continue
tispecies probiotic preparation conducted over 5 months showed to occupy the minds of researchers and that the multidisciplinary
major benefits for both distension and abdominal pain in con- approaches that have brought us thus far will be evident in meet-
junction with a stabilization of the microbiota and was completed ing the exciting challenges that lie ahead. The likelihood is that we
without any adverse effects (Kajander et al., 2008). It is also impor- have only seen the tip of the iceberg in this rapidly expanding area
tant to note the potential benefits of a treatment option with an of research.
enviable safety record and negligible toxicity profile compared to
conventional pharmacological approaches (Kopp-Hoolihan, 2001; ACKNOWLEDGMENTS
Parvez et al., 2006). Regulatory hurdles also need to be cleared with The Alimentary Pharmabiotic Centre is a research centre funded
the current classification of probiotics as food supplements limit- by Science Foundation Ireland (SFI), through the Irish Govern-
ing sanctioned use to healthy populations and functional claims ments National Development Plan. The authors and their work
over recommendations for application to the treatment of disease were supported by SFI (grant nos. 02/CE/B124 and 07/CE/B1368)
states (Marteau, 2010). Even these functional claims are the sub- and by GlaxoSmithKline. Gerard Clarke is in receipt of a
ject of controversy with agencies like the European Food Safety research grant from the American Neurogastroenterology and
Authority (EFSA) in disagreement with advocates of probiotics Motility Society (ANMS). The authors would like to thank
over what constitutes solid scientific evidence for a particular Dr. Marcela Julio for preparing the images used in this review
health claim (Guarner et al., 2011). In reality we are probably quite (http://www.facebook.com/imagenesciencia).

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 10


Grenham et al. Braingutmicrobe axis

REFERENCES Santini, D., Pasquinelli, G., Morselli- of irritable bowel syndrome: a sys- Clarke, G., Quigley, E. M., Cryan, J. F.,
Abrams, G. D., Bauer, H., and Sprinz, H. Labate, A. M., Grady, E. F., Bun- tematic review. Am. J. Gastroenterol. and Dinan, T. G. (2009b). Irritable
(1963). Influence of the normal flora nett, N. W., Collins, S. M., and Cori- 104, 10331049; quiz 1050. bowel syndrome: towards biomarker
on mucosal morphology and cellular naldesi, R. (2004). Activated mast Brint, E. K., Macsharry, J., Fanning, identification. Trends. Mol. Med. 15,
renewal in the ileum. A comparison cells in proximity to colonic nerves A., Shanahan, F., and Quigley, E. 478489.
of germ-free and conventional mice. correlate with abdominal pain in M. M. (2011). Differential expres- Clayton, E. M., Rea, M. C., Shanahan, F.,
Lab. Invest. 12, 355364. irritable bowel syndrome. Gastroen- sion of toll-like receptors in patients Quigley, E. M., Kiely, B., Hill, C., and
Adlerberth, I., and Wold, A. E. (2009). terology 126, 693702. with irritable bowel syndrome. Am. Ross, R. P. (2009). The vexed rela-
Establishment of the gut microbiota Beaver, M. H., and Wostmann, J. Gastroenterol. 106, 329336. tionship between Clostridium diffi-
in Western infants. Acta Paediatr. 98, B. (1962). Histamine and 5- Camilleri, M. (2001). Management of cile and inflammatory bowel disease:
229238. hydroxytryptamine in the intestinal the irritable bowel syndrome. Gas- an assessment of carriage in an out-
Akira, S., and Hemmi, H. (2003). tract of germ-free animals, animals troenterology 120, 652668. patient setting among patients in
Recognition of pathogen-associated harbouring one microbial species Cario, E., Gerken, G., and Podolsky, remission. Am. J. Gastroenterol. 104,
molecular patterns by TLR family. and conventional animals. Br. J. D. K. (2004). Toll-like receptor 2 11621169.
Immunol. Lett. 85, 8595. Pharmacol. Chemother. 19, 385. enhances ZO-1-associated intestinal Codling, C., OMahony, L., Shanahan, F.,
American Psychiatric Association. Bennet, R., Eriksson, M., and Nord, C. E. epithelial barrier integrity via pro- Quigley, E., and Marchesi, J. (2010).
(2000). Diagnostic criteria for (2002). The fecal microflora of 13- tein kinase C. Gastroenterology 127, A molecular analysis of fecal and
299.00 Autistic Disorder, in Diag- month-old infants during treatment 224238. mucosal bacterial communities in
nostic and Statistical Manual of with eight oral antibiotics. Infection Cash, H. L., Whitham, C. V., Behrendt, irritable bowel syndrome. Dig. Dis.
Mental Disorders. 4th, text revision 30, 158160. C. L., and Hooper, L. V. (2006). Sci. 55, 392397.
(DSM-IV-TR). Washington DC: Bercik, P., Denou, E., Collins, J., Jack- Symbiotic bacteria direct expression Collins, S. M. (2002). A case for an
American Psychiatric Association. son, W., Lu, J., Jury, J., Deng, of an intestinal bactericidal lectin. immunological basis for irritable
Andus, T., and Gross,V. (2000). Etiology Y., Blennerhassett, P., Macri, J., Science 313, 11261130. bowel syndrome. Gastroenterology
and pathophysiology of inflamma- Mccoy, K. D., Verdu, E. F., and Cenac, N., Andrews, C. N., Holzhausen, 122, 20782080.
tory bowel disease Environmental Collins, S. M. (2011). The intesti- M., Chapman, K., Cottrell, G., Collins, S. M., Denou, E., Verdu, E. F.,
factors. Hepatogastroenterology 47, nal microbiota affect central levels Andrade-Gordon, P., Steinhoff, M., and Bercik, P. (2009). The putative
2943. of brain-derived neurotropic factor Barbara, G., Beck, P., Bunnett, N. W., role of the intestinal microbiota in
Aragon, G., Graham, D. B., Borum, M., and behavior in mice. Gastroenterol- Sharkey, K. A., Ferraz, J. G. P., Shaf- the irritable bowel syndrome. Dig.
and Doman, D. B. (2010). Probi- ogy 141, 599609. fer, E., and Vergnolle, N. (2007). Role Liver Dis. 41, 850853.
otic therapy for irritable bowel syn- Berg, R. D. (1996). The indigenous for protease activity in visceral pain Corr, S. C., Gahan, C. G. M., and Hill,
drome. Gastroenterol. Hepatol. (N. gastrointestinal microflora. Trends in irritable bowel syndrome. J. Clin. C. (2007a). Impact of selected Lacto-
Y.) 6, 3944. Microbiol. 4, 430435. Invest. 117, 636647. bacillus and Bifidobacterium species
Archie, E. A., and Theis, K. R. (2011). Bischoff, S. C. (2010).Gut health: a new Chadwick, V. S., Chen, W., Shu, D., on Listeria monocytogenes infection
Animal behaviour meets microbial objective in medicine? BMC Med. 9, Paulus, B., Bethwaite, P., Tie, A., and and the mucosal immune response.
ecology. Anim. Behav. 82, 425436. 24. doi:10.1186/1741-7015-9-24 Wilson, I. (2002). Activation of the FEMS Immunol. Med. Microbiol. 50,
Bckhed, F., Ding, H., Wang, T., Bloom, S. M., Bijanki, V. N., Nava, mucosal immune system in irrita- 380388.
Hooper, L. V., Koh, G. Y., Nagy, G. M., Sun, L., Malvin, N. P., ble bowel syndrome. Gastroenterol- Corr, S. C., Li, Y., Riedel, C. U., OToole,
A., Semenkovich, C. F., and Gor- Donermeyer, D. L., Dunne, W. ogy 122, 17781783. P. W., Hill, C., and Gahan, C. G.
don, J. I. (2004). The gut micro- M. Jr., Allen, P. M., and Stap- Claesson, M. J., Cusack, S., OSullivan, M. (2007b). Bacteriocin production
biota as an environmental fac- penbeck, T. S. (2011). Commen- O., Greene-Diniz, R., De Weerd, H., as a mechanism for the antiinfec-
tor that regulates fat storage. sal Bacteroides species induce coli- Flannery, E., Marchesi, J. R., Falush, tive activity of Lactobacillus salivar-
Proc. Natl. Acad. Sci. U.S.A. 101, tis in host-genotype-specific fashion D., Dinan, T., Fitzgerald, G., Stan- ius UCC118. Proc. Natl. Acad. Sci.
1571815723. in a mouse model of inflammatory ton, C., Van Sinderen, D., OConnor, U.S.A. 104, 76177621.
Bckhed, F., Manchester, J. K., bowel disease. Cell Host Microbe 9, M., Harnedy, N., OConnor, K., Cryan, J. F., and OMahony, S. M.
Semenkovich, C. F., and Gor- 390403. Henry, C., OMahony, D., Fitzger- (2011). The microbiome-gut-brain
don, J. I. (2007). Mechanisms Bloom, S. R., Kuhajda, F. P., Laher, I., ald, A. P., Shanahan, F., Twomey, C., axis: from bowel to behavior. Neu-
underlying the resistance to diet- Pi-Sunyer, X., Ronnett, G. V., Tan, T. Hill, C., Ross, R. P., and OToole, P. rogastroenterol. Motil. 23, 187192.
induced obesity in germ-free mice. M. M., and Weigle, D. S. (2008). The W. (2011). Composition, variability, Demaude, J., Salvador-Cartier, C., Fio-
Proc. Natl. Acad. Sci. U.S.A. 104, obesity epidemic pharmacological and temporal stability of the intesti- ramonti, J., Ferrier, L., and Bueno,
979984. challenges. Mol. Interv. 8, 8298. nal microbiota of the elderly. Proc. L. (2006). Phenotypic changes in
Bailey, M. T., and Coe, C. L. (1999). Blumberg, R. S., and Strober, W. (2001). Natl. Acad. Sci. U.S.A. 108(Suppl. 1), colonocytes following acute stress
Maternal separation disrupts the Prospects for research in inflamma- 45864591. or activation of mast cells in mice:
integrity of the intestinal microflora tory bowel disease. J. Am. Med. Assoc. Clarke, G., Fitzgerald, P., Hennessy, A. implications for delayed epithe-
in infant rhesus monkeys. Dev. Psy- 285, 643647. A., Cassidy, E. M., Quigley, E. M., lial barrier dysfunction. Gut 55,
chobiol. 35, 146155. Bolte, E. R. (1998). Autism and Clostrid- Ross, P., Stanton, C., Cryan, J. F., 655661.
Bailey, M. T., Engler, H., and Sheridan, ium tetani. Med. Hypotheses 51, and Dinan, T. G. (2010). Marked Desbonnet, L., Garrett, L., Clarke, G.,
J. F. (2006). Stress induces the 133144. elevations in pro-inflammatory Bienenstock, J., and Dinan, T. G.
translocation of cutaneous and Bravo, J. A., Forsythe, P., Chew, M. polyunsaturated fatty acid metabo- (2008). The probiotic Bifidobac-
gastrointestinal microflora to sec- V., Escaravage, E., Savignac, H. M., lites in females with irritable teria infantis: an assessment of
ondary lymphoid organs of C57BL/6 Dinan, T. G., Bienenstock, J., and bowel syndrome. J. Lipid Res. 51, potential antidepressant properties
mice. J. Neuroimmunol. 171, Cryan, J. F. (2011). Ingestion of Lac- 11861192. in the rat. J. Psychiatr. Res. 43,
2937. tobacillus strain regulates emotional Clarke, G., OMahony, S. M., Hen- 164174.
Bailey, M. T., Lubach, G. R., and Coe, behavior and central GABA recep- nessy, A. A., Ross, P., Stanton, C., Desbonnet, L., Garrett, L., Clarke,
C. L. (2004). Prenatal stress alters tor expression in a mouse via the Cryan, J. F., and Dinan, T. G. G., Kiely, B., Cryan, J. F., and
bacterial colonization of the gut in vagus nerve. Proc. Natl. Acad. Sci. (2009a). Chain reactions: early-life Dinan, T. G. (2010). Effects of
infant monkeys. J. Pediatr. Gastroen- doi: 10.1073/pnas.1102999108 stress alters the metabolic profile of the probiotic Bifidobacterium infan-
terol. Nutr. 38, 414421. Brenner, D. M., Moeller, M. J., Chey, W. plasma polyunsaturated fatty acids tis in the maternal separation model
Barbara, G., Stanghellini, V., De Gior- D., and Schoenfeld, P. S. (2009). The in adulthood. Behav. Brain Res. 205, of depression. Neuroscience 170,
gio, R., Cremon, C., Cottrell, G. S., utility of probiotics in the treatment 319321. 11791188.

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 11


Grenham et al. Braingutmicrobe axis

Dinan, T. G., Clarke, G., Quigley, E. M., H., and Marlowe, E. M. (2002). Gas- Gwee, K., Collins, S., Read, N., Raj- patients: a controlled 6-month inter-
Scott, L. V., Shanahan, F., Cryan, J., trointestinal microflora studies in nakova, A., Deng, Y., Graham, J., vention. Aliment. Pharmacol. Ther.
Cooney, J., and Keeling, P. W. (2008). late-onset autism. Clin. Infect. Dis. Mckendrick, M., and Moochhala, 22, 387394.
Enhanced cholinergic-mediated 35, S6. S. (2003). Increased rectal mucosal Kajander, K., Myllyluoma, E., Rajilic-
increase in the pro-inflammatory Forsythe, P., Sudo, N., Dinan, T., Tay- expression of interleukin 1 in Stojanovic, M., Kyronpalo, S., Ras-
cytokine IL-6 in irritable bowel lor, V. H., and Bienenstock, J. (2010). recently acquired post-infectious mussen, M., Jarvenpaa, S., Zoe-
syndrome: role of muscarinic Mood and gut feelings. Brain Behav. irritable bowel syndrome. Gut 52, tendal, E. G., De Vos, W. M., Vapaat-
receptors. Am. J. Gastroenterol. 103, Immun. 24, 916. 523. alo, H., and Korpela, R. (2008). Clin-
25702576. Frank, D. N., St Amand, A. L., Gwee, K., Leong, Y., Graham, C., Mck- ical trial: multispecies probiotic sup-
Dinan, T. G., Quigley, E. M., Ahmed, Feldman, R. A., Boedeker, E. C., endrick, M., Collins, S., Walters, S., plementation alleviates the symp-
S. M., Scully, P., OBrien, S., Harpaz, N., and Pace, N. R. (2007). Underwood, J., and Read, N. (1999). toms of irritable bowel syndrome
OMahony, L., OMahony, S., Molecular-phylogenetic characteri- The role of psychological and bio- and stabilizes intestinal microbiota.
Shanahan, F., and Keeling, zation of microbial community logical factors in postinfective gut Aliment. Pharmacol. Ther. 27, 4857.
P. W. (2006). Hypothalamic- imbalances in human inflammatory dysfunction. Gut 44, 400. Kassinen, A., Krogius-Kurikka, L.,
pituitary-gut axis dysregulation in bowel diseases. Proc. Natl. Acad. Sci. Haslam, D. W., and James, W. P. Mkivuokko, H., Rinttil, T., Paulin,
irritable bowel syndrome: plasma U.S.A. 104, 1378013785. T. (2005). Obesity. Lancet 366, L., Corander, J., Malinen, E., Apa-
cytokines as a potential biomarker? Freestone, P. P., Williams, P. H., Haigh, 11971209. jalahti, J., and Palva, A. (2007). The
Gastroenterology 130, 304311. R. D., Maggs, A. F., Neal, C. P., and Heijtz, R. D., Wang, S., Anuar, F., Qian, fecal microbiota of irritable bowel
Donskey, C. J., Hujer, A. M., Das, S. M., Lyte, M. (2002). Growth stimulation Y., Bjorkholm, B., Samuelsson, A., syndrome patients differs signifi-
Pultz, N. J., Bonomo, R. A., and Rice, of intestinal commensal Escherichia Hibberd, M. L., Forssberg, H., and cantly from that of healthy subjects.
L. B. (2003). Use of denaturing gra- coli by catecholamines: a possi- Pettersson, S. (2011). Normal gut Gastroenterology 133, 2433.
dient gel electrophoresis for analysis ble contributory factor in trauma- microbiota modulates brain devel- Kerckhoffs, A. P. M., Samsom, M., Van
of the stool microbiota of hospital- induced sepsis. Shock 18, 465470. opment and behavior. Proc. Natl. Der Rest, M. E., De Vogel, J., Knol,
ized patients. J. Microbiol. Methods Freestone, P. P. E., Haigh, R. D., Acad. Sci. U.S.A. 108, 30473052. J., Ben-Amor, K., and Akkermans, L.
54, 249256. Williams, P. H., and Lyte, M. Heuvelin, E., Lebreton, C., Bichara, M., M. A. (2009). Lower Bifidobacteria
Drossman, D. A., Camilleri, M., Mayer, (2003). Involvement of enterobactin Cerf-Bensussan, N., and Heyman, counts in both duodenal mucosa-
E. A., and Whitehead, W. E. (2002). in norepinephrine-mediated iron M. (2009). A Bifidobacterium probi- associated and fecal microbiota in
AGA technical review on irritable supply from transferrin to entero- otic strain and its soluble factors alle- irritable bowel syndrome patients.
bowel syndrome. Gastroenterology haemorrhagic Escherichia coli. FEMS viate chloride secretion by human World J. Gastroenterol. 15, 2887.
123, 21082131. Microbiol. Lett. 222, 3943. intestinal epithelial cells. J. Nutr. 140, Kiliaan, A. J., Saunders, P. R., Bijlsma,
Dunlop, S. P., Jenkins, D., Neal, K. R., Gareau, M. G., Wine, E., Rodrigues, 711. P. B., Berin, M. C., Taminiau, J.
and Spiller, R. C. (2003). Relative D. M., Cho, J. H., Whary, M. Hong, K. S., Kang, H. W., Im, J. P., Ji, G. A., Groot, J. A., and Perdue, M.
importance of enterochromaffin cell T., Philpott, D. J., Macqueen, G., E., Kim, S. G., Jung, H. C., Song, I. S., H. (1998). Stress stimulates transep-
hyperplasia, anxiety, and depression and Sherman, P. M. (2011). Bacte- and Kim, J. S. (2009). Effect of probi- ithelial macromolecular uptake in
in postinfectious IBS. Gastroenterol- rial infection causes stress-induced otics on symptoms in Korean adults rat jejunum. Am. J. Physiol. 275,
ogy 125, 16511659. memory dysfunction in mice. Gut with irritable bowel syndrome. Gut G1037G1044.
Eckburg, P. B., Bik, E. M., Bernstein, 60, 307317. Liver 3, 101107. Kopp-Hoolihan, L. (2001). Prophylac-
C. N., Purdom, E., Dethlefsen, L., Gill, S. R., Pop, M., Deboy, R. T., Eck- Hong, Y. S., Hong, K. S., Park, M. H., tic and therapeutic uses of probi-
Sargent, M., Gill, S. R., Nelson, burg, P. B., Turnbaugh, P. J., Samuel, Ahn, Y. T., Lee, J. H., Huh, C. S., otics: a review. J. Am. Diet. Assoc. 101,
K. E., and Relman, D. A. (2005). B. S., Gordon, J. I., Relman, D. A., Lee, J., Kim, I. K., Hwang, G. S., 229238; quiz 239241.
Diversity of the human intesti- Fraser-Liggett, C. M., and Nelson, K. and Kim, J. S. (2011). Metabonomic Kurokawa, K., Itoh, T., Kuwahara,
nal microbial flora. Science 308, E. (2006). Metagenomic analysis of understanding of probiotic effects T., Oshima, K., Toh, H., Toy-
16351638. the human distal gut microbiome. in humans with irritable bowel syn- oda, A., Takami, H., Morita, H.,
Ewaschuk, J. B., Diaz, H., Meddings, Science 312, 13551359. drome. J. Clin. Gastroenterol. 45, Sharma, V. K., and Srivastava, T. P.
L., Diederichs, B., Dmytrash, A., Glaister, J. (1973). Factors affecting the 415425. (2007). Comparative metagenomics
Backer, J., Looijer-Van Langen, M., lymphoid cells in the small intestinal Hooper, L. V., Stappenbeck, T. S., revealed commonly enriched gene
and Madsen, K. L. (2008). Secreted epithelium of the mouse. Int. Arch. Hong, C. V., and Gordon, J. I. sets in human gut microbiomes.
bioactive factors from Bifidobac- Allergy Immunol. 45, 719730. (2003). Angiogenins: a new class DNA Res. 14, 169.
terium infantis enhance epithelial Gordon, H. A. (1959). Morphological of microbicidal proteins involved in Ley, R. E., Bckhed, F., Turnbaugh, P.,
cell barrier function. Am. J. Phys- and physiological characterization innate immunity. Nat. Immunol. 4, Lozupone, C. A., Knight, R. D., and
iol. Gastrointest. Liver Physiol. 295, of germfree life. Ann. N. Y. Acad. Sci. 269273. Gordon, J. I. (2005). Obesity alters
G1025G1034. 78, 208220. Hooper, L. V., Wong, M. H., Thelin, A., gut microbial ecology. Proc. Natl.
Finegold, S. M. (2008). Therapy and Gordon, H. A., and Bruckner-Kardoss, Hansson, L., Falk, P. G., and Gordon, Acad. Sci. U.S.A. 102, 1107011075.
epidemiology of autism-clostridial E. (1961). Effect of normal micro- J. I. (2001). Molecular analysis of Ley, R. E., Peterson, D. A., and Gordon, J.
spores as key elements. Med. bial flora on intestinal surface area. commensal host-microbial relation- I. (2006a). Ecological and evolution-
Hypotheses 70, 508511. Am. J. Physiol. 201, 175. ships in the intestine. Science 291, ary forces shaping microbial diver-
Finegold, S. M., Dowd, S. E., Greer, J. B., and OKeefe, S. J. 881884. sity in the human intestine. Cell 124,
Gontcharova, V., Liu, C., Hen- (2011). Microbial induction of Joossens, M., Huys, G., Cnockaert, M., 837848.
ley, K. E., Wolcott, R. D., Youn, E., immunity, inflammation, and De Preter, V., Verbeke, K., Rutgeerts, Ley, R. E., Turnbaugh, P. J., Klein, S.,
Summanen, P. H., Granpeesheh, cancer. Front. Physiol. 1:168. P., Vandamme, P., and Vermeire, and Gordon, J. I. (2006b). Micro-
D., Dixon, D., Liu, M., Molitoris, doi:10.3389/fphys.2010.00168 S. (2011). Dysbiosis of the faecal bial ecology: human gut microbes
D. R., and Green Iii, J. A. (2010). Guarner, F., Ellen Sanders, M., Gibson, microbiota in patients with Crohns associated with obesity. Nature 444,
Pyrosequencing study of fecal G., Klaenhammer, T., Cabana, M., disease and their unaffected rela- 10221023.
microflora of autistic and control Scott, K., Reid, G., Delzenne, N. M., tives. Gut 60, 631637. Lichtenstein, G. R. (2000). Chemokines
children. Anaerobe 16, 444453. Fahey, G. C., and Hill, C. (2011). Kajander, K., Hatakka, K., Poussa, T., and cytokines in inflammatory
Finegold, S. M., Molitoris, D., Song, Y., Probiotic and prebiotic claims in Farkkila, M., and Korpela, R. (2005). bowel disease and their application
Liu, C., Vaisanen, M. L., Bolte, E., Europe: seeking a clear roadmap. Br. A probiotic mixture alleviates symp- to disease treatment. Curr. Opin.
Mcteague, M., Sandler, R., Wexler, J. Nutr. 1, 13. toms in irritable bowel syndrome Gastroenterol. 16, 8388.

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 12


Grenham et al. Braingutmicrobe axis

Liebregts, T., Adam, B., Bredack, Marshall, J. K., Thabane, M., Bor- Melgar, S., and Shanahan, F. (2010). OMahony, S. M., Marchesi, J. R.,
C., Rth, A., Heinzel, S., Lester, gaonkar, M. R., and James, C. (2007). Inflammatory bowel disease-from Scully, P., Codling, C., Ceolho, A.
S., Downie-Doyle, S., Smith, E., Postinfectious irritable bowel syn- mechanisms to treatment strategies. M., Quigley, E. M., Cryan, J. F.,
Drew, P., Talley, N. J., and Holt- drome after a food-borne outbreak Autoimmunity 43, 463477. and Dinan, T. G. (2009). Early life
mann, G. (2007). Immune acti- of acute gastroenteritis attributed to Messaoudi, M., Lalonde, R., Violle, N., stress alters behavior, immunity, and
vation in patients with irritable a viral pathogen. Clin. Gastroenterol. Javelot, H., Desor, D., Nejdi, A., microbiota in rats: implications for
bowel syndrome. Gastroenterology Hepatol. 5, 457460. Bisson, J. F., Rougeot, C., Piche- irritable bowel syndrome and psy-
132, 913920. Marshall, J. K., Thabane, M., Garg, A. lin, M., Cazaubiel, M., and Caza- chiatric illnesses. Biol. Psychiatry 65,
Lupp, C., Robertson, M. L., Wick- X., Clark, W. F., Salvadori, M., and ubiel, J. M. (2011). Assessment of 263267.
ham, M. E., Sekirov, I., Champion, Collins, S. M. (2006). Incidence and psychotropic-like properties of a Palmer, C., Bik, E. M., Digiulio, D.
O. L., Gaynor, E. C., and Fin- epidemiology of irritable bowel syn- probiotic formulation (Lactobacil- B., Relman, D. A., and Brown,
lay, B. B. (2007). Host-mediated drome after a large waterborne out- lus helveticus R0052 and Bifidobac- P. O. (2007). Development of the
inflammation disrupts the intestinal break of bacterial dysentery. Gas- terium longum R0175) in rats and human infant intestinal microbiota.
microbiota and promotes the over- troenterology 131, 445450. human subjects. Br. J. Nutr. 105, PLoS Biol. 5, e177. doi:10.1371/jour-
growth of Enterobacteriaceae. Cell Marteau, P. (2010). Probiotics in func- 755764. nal.pbio.0050177
Host Microbe 2, 204. tional intestinal disorders and IBS: Mikkelsen, H. B., Garbarsch, C., Tra- Pandol, S. J. (2010). Integrating the gas-
Lyra, A., Krogius-Kurikka, L., Nikkila, J., proof of action and dissecting num-Jensen, J., and Thuneberg, L. trointestinal sciences. Front. Physiol.
Malinen, E., Kajander, K., Kurikka, the multiple mechanisms. Gut 59, (2004). Macrophages in the small 1:19. doi:10.3389/fphys.2010.00019
K., Korpela, R., and Palva, A. 285286. intestinal muscularis externa of Parracho, H. M., Bingham, M. O., Gib-
(2010). Effect of a multispecies Matsumoto, S., Setoyama, H., and Ume- embryos, newborn and adult germ- son, G. R., and Mccartney, A. L.
probiotic supplement on quan- saki, Y. (1992). Differential induc- free mice. J. Mol. Histol. 35, 377387. (2005). Differences between the gut
tity of irritable bowel syndrome- tion of major histocompatibility Million, M., Maraninchi, M., Henry, microflora of children with autis-
related intestinal microbial phylo- complex molecules on mouse intes- M., Armougom, F., Richet, H., Car- tic spectrum disorders and that of
types. BMC Gastroenterol. 10, 110. tine by bacterial colonization. Gas- rieri, P., Valero, R., Raccah, D., healthy children. J. Med. Microbiol.
doi:10.1186/1471-230X-10-110 troenterology 103, 1777. Vialettes, B., and Raoult, D. (2011). 54, 987991.
Lyte, M., Li, W., Opitz, N., Gaykema, R., Mtt, J., Maunuksela, L., Kajander, K., Obesity-associated gut microbiota Parvez, S., Malik, K. A., Ah Kang, S.,
and Goehler, L. E. (2006). Induction Palva, A., Korpela, R., Kassinen, A., is enriched in Lactobacillus reuteri and Kim, H. Y. (2006). Probiotics
of anxiety-like behavior in mice dur- and Saarela, M. (2005). Composi- and depleted in Bifidobacterium and their fermented food products
ing the initial stages of infection with tion and temporal stability of gas- animalis and Methanobrevibacter are beneficial for health. J. Appl.
the agent of murine colonic hyper- trointestinal microbiota in irritable smithii. Int. J. Obes. (Lond.). doi: Microbiol. 100, 11711185.
plasia Citrobacter rodentium. Physiol. bowel syndrome a longitudinal 10.1038/ijo.2011.153 Podolsky, D. K. (2002). Inflammatory
Behav. 89, 350357. study in IBS and control subjects. Moayyedi, P., Ford, A. C., Talley, N. J., bowel disease. N. Engl. J. Med. 347,
MacDonald, T. T., and Monteleone, FEMS Immunol. Med. Microbiol. 43, Cremonini, F., Foxx-Orenstein, A. E., 417429.
G. (2005). Immunity, inflammation, 213222. Brandt, L. J., and Quigley, E. M. Qin, J., Li, R., Raes, J., Arumugam,
and allergy in the gut. Science 307, Mawdsley, J. E., and Rampton, D. S. (2008). The efficacy of probiotics in M., Burgdorf, K. S., Manichanh, C.,
1920. (2006). The role of psychological the treatment of irritable bowel syn- Nielsen, T., Pons, N., Levenez, F.,
Macfarlane, G., Blackett, K., Nakayama, stress in inflammatory bowel dis- drome: a systematic review. Gut 59, Yamada, T., Mende, D. R., Li, J., Xu,
T., Steed, H., and Macfarlane, S. ease. Neuroimmunomodulation 13, 325332. J., Li, S., Li, D., Cao, J., Wang, B.,
(2009). The gut microbiota in 327336. Moreau, M. C., Ducluzeau, R., Guy- Liang, H., Zheng, H., Xie, Y., Tap,
inflammatory bowel disease. Curr. Mayer, E. A. (2011). Gut feelings: the Grand, D., and Muller, M. C. (1978). J., Lepage, P., Bertalan, M., Batto, J.-
Pharm. Des. 15, 15281536. emerging biology of gut-brain com- Increase in the population of duode- M., Hansen, T., Le Paslier, D., Lin-
Macfarlane, S., and Macfarlane, G. T. munication. Nat. Rev. Neurosci. 12, nal immunoglobulin a plasmocytes neberg, A., Nielsen, H. B., Pelletier,
(2003). Regulation of short-chain 453466. in axenic mice associated with differ- E., Renault, P., Sicheritz-Ponten, T.,
fatty acid production. Proc. Nutri- Mayer, L. (2003). Mucosal immunity. ent living or dead bacterial strains of Turner, K., Zhu, H.,Yu, C., Li, S., Jian,
tion Soc. 62, 6772. Pediatrics 111, 15951600. intestinal origin. Infect. Immun. 21, M., Zhou, Y., Li, Y., Zhang, X., Li, S.,
Mackie, R. I., Sghir, A., and Gaskins, Mazmanian, S. K., Liu, C. H., Tziana- 532539. Qin, N., Yang, H., Wang, J., Brunak,
H. R. (1999). Developmental micro- bos, A. O., and Kasper, D. L. (2005). Neufeld, K. M., Kang, N., Bienenstock, S., Dore, J., Guarner, F., Kristiansen,
bial ecology of the neonatal gastroin- An immunomodulatory molecule of J., and Foster, J. A. (2011). Reduced K., Pedersen, O., Parkhill, J., Weis-
testinal tract. Am. J. Clin. Nutr. 69, symbiotic bacteria directs matura- anxiety-like behavior and central senbach, J., Bork, P., Ehrlich, S. D.,
1035S1045S. tion of the host immune system. Cell neurochemical change in germ-free and Wang, J. (2010). A human gut
Malinen, E., Rinttila, T., Kajander, K., 122, 107118. mice. Neurogastroenterol. Motil. 23, microbial gene catalogue established
Matto, J., Kassinen, A., Krogius, L., McCracken, V. J., and Lorenz, R. G. 255264. by metagenomic sequencing. Nature
Saarela, M., Korpela, R., and Palva, A. (2001). The gastrointestinal ecosys- OHara, A. M., and Shanahan, F. (2006). 464, 5965.
(2005). Analysis of the fecal micro- tem: a precarious alliance among The gut flora as a forgotten organ. Quigley, E. M. (2007). Bacterial flora
biota of irritable bowel syndrome epithelium, immunity and micro- EMBO Rep. 7, 688693. in irritable bowel syndrome: role
patients and healthy controls with biota. Cell. Microbiol. 3, 111. OMahony, L., Mccarthy, J., Kelly, P., in pathophysiology, implications for
real-time PCR. Am. J. Gastroenterol. McKernan, D. P., Fitzgerald, P., Dinan, Hurley, G., Luo, F., Chen, K., management. J. Dig. Dis. 8, 27.
100, 373382. T. G., and Cryan, J. F. (2010). OSullivan, G. C., Kiely, B., Collins, J. Quigley, E. M. (2008). Probiotics in
Mndar, R., and Mikelsaar, M. (1996). The probiotic Bifidobacterium infan- K., Shanahan, F., and Quigley, E. M. functional gastrointestinal disor-
Transmission of mothers microflora tis 35624 displays visceral antinoci- (2005). Lactobacillus and Bifidobac- ders: what are the facts? Curr. Opin.
to the newborn at birth. Neonatology ceptive effects in the rat. Neuro- terium in irritable bowel syndrome: Pharmacol. 8, 704708.
69, 3035. gastroenterol. Motil. 22, 10291035, symptom responses and relationship Quigley, E. M. (2009). Do patients with
Mangell, P., Nejdfors, P., Wang, M., e1268. to cytokine profiles. Gastroenterol- functional gastrointestinal disorders
Ahrne, S., Westrom, B., Thorlacius, McKernan, D. P., Gaszner, G., Quigley, ogy 128, 541551. have an altered gut flora? Therap.
H., and Jeppsson, B. (2002). Lac- E. M., Cryan, J. F., and Dinan, T. G. OMahony, S. M., Hyland, N. P., Dinan, Adv. Gastroenterol. 2, 2330.
tobacillus plantarum 299v inhibits (2011). Altered peripheral toll-like T. G., and Cryan, J. F. (2011). Mater- Quigley, E. M. M. (2011). Antibiotics
Escherichia coli-induced intestinal receptor responses in the irritable nal separation as a model of brain- in irritable bowel syndrome: a novel
permeability. Dig. Dis. Sci. 47, bowel syndrome. Aliment. Pharma- gut axis dysfunction. Psychopharma- approach to a challenging disorder.
511516. col. Ther. 33, 10451052. cology (Berl.) 214, 7188. Clin. Investig. (Lond.) 1, 479482.

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 13


Grenham et al. Braingutmicrobe axis

RajilicStojanovic, M., Biagi, E., Heilig, Culture-independent analyses of Spiller, R., and Garsed, K. (2009). intestinal host-microbial interface.
H. G. H. J., Kajander, K., Kekko- temporal variation of the dominant Postinfectious irritable bowel Proc. Natl. Acad. Sci. U.S.A. 105,
nen, R. A., Tims, S., and De Vos, W. fecal microbiota and targeted bacte- syndrome. Gastroenterology 136, 2085820863.
M. (2011). Global and deep molecu- rial subgroups in Crohns disease. J. 19791988. Verdu, E. F., Bercik, P., Verma-Gandhu,
lar analysis of microbiota signatures Clin. Microbiol. 44, 39803988. Spiller, R. C., Jenkins, D., Thornley, J. M., Huang, X. X., Blennerhassett,
in fecal samples from patients with Schellekens, H., Dinan, T. G., and P., Hebden, J. M., Wright, T., Skin- P., Jackson, W., Mao, Y., Wang, L.,
irritable bowel syndrome. Gastroen- Cryan, J. F. (2010). Lean mean ner, M., and Neal, K. R. (2000). Rochat, F., and Collins, S. M. (2006).
terology 141, 17921801. fat reducing ghrelin machine: Increased rectal mucosal enteroen- Specific probiotic therapy attenuates
Rakoff-Nahoum, S., Paglino, J., Eslami- hypothalamic ghrelin and ghre- docrine cells, T lymphocytes, and antibiotic induced visceral hyper-
Varzaneh, F., Edberg, S., and Medzhi- lin receptors as therapeutic targets increased gut permeability following sensitivity in mice. Gut 55, 182190.
tov, R. (2004). Recognition of com- in obesity. Neuropharmacology 58, acute Campylobacter enteritis and in Walker, A., Sanderson, J., Churcher, C.,
mensal microflora by toll-like recep- 216. post-dysenteric irritable bowel syn- Parkes, G., Hudspith, B., Rayment,
tors is required for intestinal home- Scully, P., McKernan, D. P., Keo- drome. Gut 47, 804811. N., Brostoff, J., Parkhill, J., Dougan,
ostasis. Cell 118, 229241. hane, J., Groeger, D., Shanahan, Stappenbeck, T. S., Hooper, L. V., and G., and Petrovska, L. (2011). High-
Reber, S. O. (2011). Stress and animal F., Dinan, T. G., and Quigley, E. Gordon, J. I. (2002). Developmental throughput clone library analysis of
models of inflammatory bowel dis- M. (2010). Plasma cytokine pro- regulation of intestinal angiogenesis the mucosa-associated microbiota
ease An update on the role of files in females with irritable bowel by indigenous microbes via Paneth reveals dysbiosis and differences
the hypothalamopituitaryadrenal syndrome and extra-intestinal co- cells. Proc. Natl. Acad. Sci. U.S.A. 99, between inflamed and non-inflamed
axis. Psychoneuroendocrinology. doi: morbidity. Am. J. Gastroenterol. 105, 15451. regions of the intestine in inflamma-
10.1016/j.psyneuen.2011.05.014 22352243. Sudo, N., Chida, Y., Aiba, Y., Sonoda, tory bowel disease. BMC Microbiol.
Rhee, S. H., Pothoulakis, C., and Mayer, Sekirov, I., Russell, S. L., Antunes, L. J., Oyama, N., Yu, X. N., Kubo, 11, 7. doi:10.1186/1471-2180-11-7
E. A. (2009). Principles and clini- C. M., and Finlay, B. B. (2010). Gut C., and Koga, Y. (2004). Postnatal Whorwell, P. J. (2009). Do probiotics
cal implications of the braingut microbiota in health and disease. microbial colonization programs the improve symptoms in patients with
enteric microbiota axis. Nat. Rev. Physiol. Rev. 90, 859904. hypothalamic-pituitary-adrenal sys- irritable bowel syndrome? Therap.
Gastroenterol. Hepatol. 6, 306314. Shanahan, F. (2002). The host-microbe tem for stress response in mice. J. Adv. Gastroenterol. 2, 3744.
Rodrguez, L. A. G., and Ruigmez, A. interface within the gut. Best Pract. Physiol. (Lond.) 558, 263275. Willing, B., Halfvarson, J., Dicksved,
(1999). Increased risk of irritable Res. Clin. Gastroenterol. 16, 915931. Sumi, Y., Miyakawa, M., Kanzaki, M., J., Rosenquist, M., Jrnerot, G.,
bowel syndrome after bacterial gas- Shanahan, F. (2004). Probiotics in and Kotake, Y. (1977). Vitamin B-6 Engstrand, L., Tysk, C., and Jans-
troenteritis: cohort study. BMJ 318, inflammatory bowel disease thera- deficiency in germfree rats. J. Nutr. son, J. K. (2009). Twin stud-
565. peutic rationale and role. Adv. Drug 107, 1707. ies reveal specific imbalances in
Rousseaux, C., Thuru, X., Gelot, A., Deliv. Rev. 56, 809818. Tana, C., Umesaki, Y., Imaoka, A., the mucosa-associated microbiota
Barnich, N., Neut, C., Dubuquoy, Shanahan, F. (2005). Physiological basis Handa, T., Kanazawa, M., and of patients with ileal Crohns dis-
L., Dubuquoy, C., Merour, E., for novel drug therapies used to Fukudo, S. (2010). Altered profiles ease. Inflamm. Bowel Dis. 15,
Geboes, K., Chamaillard, M., Ouwe- treat the inflammatory bowel dis- of intestinal microbiota and organic 653660.
hand, A., Leyer, G., Carcano, D., eases I. Pathophysiological basis and acids may be the origin of symptoms Woodmansey, E. J. (2007). Intestinal
Colombel, J. F., Ardid, D., and prospects for probiotic therapy in in irritable bowel syndrome. Neu- bacteria and ageing. J. Appl. Micro-
Desreumaux, P. (2007). Lactobacil- inflammatory bowel disease. Am. J. rogastroenterol. Motil. 22, 512519, biol. 102, 11781186.
lus acidophilus modulates intesti- Physiol. Gastrointest. Liver Physiol. e114e115. Wostmann, B., and Bruckner-Kardoss,
nal pain and induces opioid and 288, G417G421. Thompson, W. G., Longstreth, G. F., E. (1959). Development of cecal dis-
cannabinoid receptors. Nat. Med. 13, Shanahan, F. (2010a). Gut microbes: Drossman, D. A., Heaton, K. W., tention in germ-free baby rats. Am.
3537. from bugs to drugs. Am. J. Gastroen- Irvine, E. J., and Mller-Lissner, S. J. Physiol. 197, 13451346.
Ruddick, J. P., Evans, A. K., Nutt, D. J., terol. 105, 275279. A. (1999). Functional bowel disor- Wostmann, B. S. (1981). The germfree
Lightman, S. L., Rook, G. A., and Shanahan, F. (2010b). Probiotics in ders and functional abdominal pain. animal in nutritional studies. Annu.
Lowry, C. A. (2006). Tryptophan perspective. Gastroenterology 139, Gut 45, II43II47. Rev. Nutr. 1, 257279.
metabolism in the central nervous 18081812. Turnbaugh, P. J., Ley, R. E., Mahowald, Wostmann, B. S., Larkin, C., Moriarty,
system: medical implications. Expert Smith, K., Mccoy, K. D., and Macpher- M. A., Magrini, V., Mardis, E. R., A., and Bruckner-Kardoss, E. (1983).
Rev. Mol. Med. 8, 127. son, A. J. (2007). Use of axenic and Gordon, J. I. (2006). An obesity- Dietary intake, energy metabolism,
Samuel, B. S., Shaito, A., Motoike, T., animals in studying the adapta- associated gut microbiome with and excretory losses of adult male
Rey, F. E., Backhed, F., Manches- tion of mammals to their com- increased capacity for energy har- germfree Wistar rats. Lab. Anim. Sci.
ter, J. K., Hammer, R. E., Williams, mensal intestinal microbiota. Semin. vest. Nature 444, 10271031. 33, 46.
S. C., Crowley, J., Yanagisawa, M., Immunol. 19, 5969. Ulevitch, R. J. (1999). Endotoxin opens Wu, S. V., and Hui, H. (2011). Treat
and Gordon, J. I. (2008). Effects Sobko, T., Reinders, C., Norin, E., the Tollgates to innate immunity. your bug right. Front. Physiol. 2:9.
of the gut microbiota on host adi- Midtvedt, T., Gustafsson, L. E., and Nat. Med. 5, 144145. doi:10.3389/fphys.2011.00009
posity are modulated by the short- Lundberg, J. O. (2004). Gastroin- Umesaki, Y., Okada, Y., Matsumoto, Xu, J., Mahowald, M. A., Ley, R.
chain fatty-acid binding G protein- testinal nitric oxide generation in S., Imaoka, A., and Setoyama, E., Lozupone, C. A., Hamady,
coupled receptor, Gpr41. Proc. Natl. germ-free and conventional rats. H. (1995). Segmented filamentous M., Martens, E. C., Henrissat, B.,
Acad. Sci. U.S.A. 105, 1676716772. Am. J. Physiol. Gastrointest. Liver bacteria are indigenous intestinal Coutinho, P. M., Minx, P., Latreille,
Sandler, R. H., Finegold, S. M., Bolte, E. Physiol. 287, G993. bacteria that activate intraepithe- P., Cordum, H., Van Brunt, A., Kim,
R., Buchanan, C. P., Maxwell, A. P., Sokol, H., Seksik, P., Rigottier-Gois, L., lial lymphocytes and induce MHC K., Fulton, R. S., Fulton, L. A.,
Visnen, M.-L., Nelson, M. N., and Lay, C., Lepage, P., Podglajen, I., class II molecules and fucosyl asialo Clifton, S. W., Wilson, R. K., Knight,
Wexler, H. M. (2000). Short-term Marteau, P., and Dor, J. (2006). GM1 glycolipids on the small intesti- R. D., and Gordon, J. I. (2007).
benefit from oral vancomycin treat- Specificities of the fecal micro- nal epithelial cells in the ex-germ- Evolution of symbiotic bacteria in
ment of regressive-onset autism. J. biota in inflammatory bowel disease. free mouse. Microbiol. Immunol. 39, the distal human intestine. PLoS
Child Neurol. 15, 429435. Inflamm. Bowel Dis. 12, 106111. 555562. Biol. 5, e156. doi:10.1371/jour-
Sarna, S. K. (2011). Lessons learnt from Song, Y., Liu, C., and Finegold, S. M. Vaishnava, S., Behrendt, C. L., Ismail, nal.pbio.0050156
post-infectious IBS. Front. Physiol. (2004). Real-time PCR quantitation A. S., Eckmann, L., and Hooper, L. Zareie, M., Johnson-Henry, K., Jury, J.,
2:49. doi:10.3389/fphys.2011.00049 of clostridia in feces of autistic chil- V. (2008). Paneth cells directly Yang, P. C., Ngan, B. Y., Mckay, D. M.,
Scanlan, P. D., Shanahan, F., OMahony, dren. Appl. Environ. Microbiol. 70, sense gut commensals and Soderholm, J. D., Perdue, M. H., and
C., and Marchesi, J. R. (2006). 64596465. maintain homeostasis at the Sherman, P. M. (2006). Probiotics

Frontiers in Physiology | Gastrointestinal Sciences December 2011 | Volume 2 | Article 94 | 14


Grenham et al. Braingutmicrobe axis

prevent bacterial translocation and commercial or financial relationships Citation: Grenham S, Clarke G, Cryan Copyright 2011 Grenham, Clarke,
improve intestinal barrier function that could be construed as a potential JF and Dinan TG (2011) Brain Cryan and Dinan. This is an open-access
in rats following chronic psycholog- conflict of interest. gutmicrobe communication in health article distributed under the terms of
ical stress. Gut 55, 15531560. and disease. Front. Physio. 2:94. doi: the Creative Commons Attribution Non
Received: 15 September 2011; paper 10.3389/fphys.2011.00094 Commercial License, which permits non-
Conflict of Interest Statement: The pending published: 13 October 2011; This article was submitted to Frontiers commercial use, distribution, and repro-
authors declare that the research was accepted: 18 November 2011; published in Gastrointestinal Sciences, a specialty of duction in other forums, provided the
conducted in the absence of any online: 07 December 2011. Frontiers in Physiology. original authors and source are credited.

www.frontiersin.org December 2011 | Volume 2 | Article 94 | 15

Вам также может понравиться