You are on page 1of 9

Neurogastroenterology & Motility

Neurogastroenterol Motil (2012) 24, 405–413 doi: 10.1111/j.1365-2982.2012.01906.x

REVIEW ARTICLE

Microbes and the gut-brain axis


P. BERCIK , S. M. COLLINS & E. F. VERDU

Farcombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada

Abstract nervous system (CNS). Although the original descrip-


Background The ‘gut-brain’ or ‘brain-gut axis’, tion of a gut-brain axis related to the modulation of
depending on whether we emphasize bottom-up or cholecystokinin secretion by bombesin,1 the concept
top-bottom pathways, is a bi-directional communica- has since then been extended to describe any interac-
tion system, comprised of neural pathways, such as tion between the gastrointestinal tract and the CNS.
the enteric nervous system (ENS), vagus, sympathetic Recently, results in animal models have generated
and spinal nerves, and humoral pathways, which in- great interest into the role of intestinal microbes as key
clude cytokines, hormones, and neuropeptides as sig- players in gut-brain communication (Fig. 1). The neu-
naling molecules. Recent evidence, mainly arising ral aspects and the role of centrally-driven pathways in
from animal models, supports a role of microbes as gut-brain axis communication have recently been
signaling components in the gut-brain axis. Aims The reviewed in detail by Mayer et al.2 and O’Mahony
purpose of this review is to summarize our current et al.3
knowledge regarding the role of microbes, including
commensals, probiotics and gastrointestinal patho-
Intestinal microbiota and gut homeostasis
gens, in bottom-up pathways of communication in the
gut-brain axis. Although this has clear implications The intestinal microbiota involves a wide diversity of
for psychiatric co-morbidity in functional and in- microbial species4 and can be considered a postnatal
flammatory conditions of the gut, the focus of this acquired organ that performs different functions for the
review will be to discuss the current evidence for a role host. Intestinal microbes have developed a mutualistic
of bacteria (commensals, probiotics, and pathogens) as relationship with its host and play a crucial role in
key modulators of gut-brain communication. Results the development of innate and adaptive immune
& Conclusions The strongest evidence for a role of responses,5,6 influence physiological systems through-
microbes as signaling components in the gut-brain axis out life by modulating gut motility, intestinal barrier
currently arises from animal studies and indicate that homeostasis,7,8 absorption of nutrients and the distri-
mechanisms of communication are likely to be multi- bution of somatic and visceral fat.9,10
ple. There is need for the concepts generated in animal The intestinal microbiota consists of a community
models to be translated to the human in the future. of bacteria that colonize the gastrointestinal tract after
birth and persist throughout adult life, and ‘transient’
bacteria, such as probiotic bacteria, which are tempo-
INTRODUCTION
rarily acquired during ingestion of certain foods. The
Clinicians and researchers have long recognized the composition of the intestinal microbiota is established
link between gastrointestinal function and the central during the first few years of life and is likely shaped by
multiple factors including maternal vertical transmis-
Address for Correspondence
sion, genetic make up of the individual, diet, medica-
Elena F. Verdu, 1280 main Street West, HSc 3N8, Hamilton, tions such as antibiotics, gastrointestinal infections
ON, L8S 4K1, Canada. and stress11–15 (Fig. 2). Until recently composition of
Tel: 905 525-9140x20051; fax: 905 522-3454; this microbial community was considered unique for
e-mail: verdue@mcmaster.ca each individual and relatively stable over time.16,17
Received: 9 January 2012
Accepted for publication: 13 Febraury 2012
However, using deep sequencing of stool samples from

Ó 2012 Blackwell Publishing Ltd 405


P. Bercik et al. Neurogastroenterology and Motility

Figure 1 The intestinal microbiome: a


complex and dynamic ecosystem that
establishes bi-directional interactions with
the host.

Figure 2 The gut-brain axis. Pathways of


communication and probiotic targets.

several hundred individuals, the European MetaHit variation in the complexity and stability of Bifidobac-
consortium study has shown that human microbiota teria and Lactobacillus populations over a 12-month
profiles can be grouped in three major bacterial period20 and significant alterations in microbiota
enterotypes dominated by Bacteroides, Prevotella composition due to environmental factors.21,22 Treat-
and Ruminococcus, respectively.18 Existence of ment with antibiotics is known to affect the intestinal
distinct enterotypes strongly associated with long- microbiota and these changes may be long lasting.
term diets has been confirmed by Wu et al., linking A study investigating the effect of a short course of
protein and animal fat with Bacteroides and con- amoxicillin showed that the microbial ecology of the
sumption of carbohydrates with Prevotella.19 This intestinal tract was severely altered for up to 6 months
indicates that despite existence of large number of after the end of antibiotic administration.23 Similarly,
bacterial strains in the human intestine there are only 1-week course of clindamycin caused changes in the
a limited number of well-balanced host-microbial Bacteroides community, and this persisted for up to
symbiotic states that might respond differently to diet 2 years.24,25 Diet is also likely to have a profound effect
and drug intake. on gut bacterial composition. Different bacterial phe-
Although the intestinal microbiota is considered notypic profiles were found in subjects consuming
relatively stable, some studies have shown a marked either a Western style or an agrarian diet on a chronic

406 Ó 2012 Blackwell Publishing Ltd


Volume 24, Number 5, May 2012 Microbiota-gut-brain axis

basis.26 Although it would be logical to expect that clearly illustrates that that the neural system can
acute changes in diet would alter the gut microbiota, detect an acute change in the gut and selectively
recent short-term experiments in healthy volunteers identifies the presence of a pathogen in the gut lumen.
using either high fat of high carbohydrate diets Studies using chronic H. pylori infection in mice
have not shown any marked changes in bacterial have shown that this pathogen alters gastric physiol-
enterotypes.19 It is thus possible that a stable micro- ogy, namely delayed gastric emptying and visceral
biota is a hallmark of health, as unstable microbial sensitivity, with up-regulation of SP and CGRP-
profiles have been reported in patients with irritable containing nerves in the stomach and the spinal
bowel syndrome and inflammatory bowel disease.27–30 cord.43,44 Furthermore, chronic H. pylori infection
leads to abnormal feeding behavior, characterized by
frequent feeding bouts but with less food consumed per
Intestinal microbiota and the gut brain axis
feeding bout than controls, which is reminiscent of
Indirect evidence for an effect of microbiota on CNS: early satiety observed in patients with functional
clinical studies The concept that gut bacteria are a dyspepsia.44 The abnormal feeding pattern was accom-
driving force for immune maturation and gut function panied by down-regulation of regulatory peptide Pro-
in the host is well accepted. The notion that bacteria opio-melanocortin (POMC) in the arcuate nucleus and
could also influence brain function and behavior is up-regulation of the pro-inflammatory cytokine TNF-a
seemingly implausible, but clinicians use on a routine in the median eminence (ME) of the hypothalamus.
basis laxatives and oral antibiotics to treat patients The ME is a part of the circumventricular organ, area of
with altered mental status due to hepatic encephalop- the brain where blood-brain barrier is relatively leaky
athy.31 Several clinical studies have also described enabling metabolites/molecules from the systemic
altered composition of gut microbiota in patients with circulation to enter the CNS. Interestingly, altered
autism32 and suggested at least short-term beneficial behavior and biochemical abnormalities persisted for
effect of antibiotic treatment,33,34 although there is no at least two months postbacterial eradication suggest-
randomized clinical trial available to date. There are ing that changes induced by chronic infection in the
also multiple reports of patients developing psychosis CNS may be long lasting or permanent.
after administration of different antibiotics.35 To our To establish a link between commensal bacteria and
knowledge, there have been no studies that character- the CNS, several experimental approaches can be
ize the gut microbiota associated with depression or undertaken. One is to compare germ-free with animals
anxiety, but earlier studies demonstrated that depres- colonized with specific pathogen flora (SPF). Sudo et al.
sion in females is associated with increased fermenta- demonstrated an abnormal HPA axis with elevated
tion of carbohydrates, indirectly implicating changes in ACTH and corticosterone levels in response to
the composition or metabolic activity of the gut restraint stress in germ-free mice, which normalized
microbiota.36,37 after colonization with commensal bacteria.45 Further-
more, germ-free mice had lower brain derived neuro-
Direct evidence for an effect of commensals and trophic factor (BDNF) levels in the cortex and
pathogens on CNS: animal studies At this point, the hippocampus. Several recent studies have compared
brunt of evidence linking microbes with behavior and behavior and brain biochemistry in germ-free and SPF
brain biochemistry comes from animal studies. Pivotal mice. Overall, using standard behavioral tests, such as
experiments performed by Lyte et al. have shown that elevated plus maze, open field and light/dark prefer-
mice display altered, anxiety-like behavior during the ence tests, germ-free mice displayed higher exploratory
early phase of acute infection with Campylobacter and lower anxiety-like behavior than SPF mice.46,47
jejuni.38 This abnormal behavior occurred within Heijtz et al. showed that compared to germ-free mice,
several hours after introduction of the intestinal path- SPF mice had higher central expression of neurotro-
ogen into the GI tract, before any significant immune phins, such as nerve growth factor (NGF) and BDNF.46
response was mounted suggesting that this was not a Furthermore, there was differential expression of
consequence of cytokine-induced sickness behavior. multiple genes involved in the secondary messenger
Subsequent studies showed that presence of C. jejuni pathways and synaptic long-term potentiation in the
or Citrobacter rodentium triggers activity of vagal hippocampus, frontal cortex and striatum. Similarly,
ascending pathways, such as nucleus tractus solitarius Neufeld et al. demonstrated increased expression of
(NTS) and the lateral parabrachial nucleus,39–41 and a NMDA receptor subunit NR2B in the central amygdala
specific activation pattern in multiple brain regions and serotonin receptor 1A (5-HT 1A) expression in the
previously implicated in anxiety-like behavior.42 This hippocampus in SPF mice compared to germ-free

Ó 2012 Blackwell Publishing Ltd 407


P. Bercik et al. Neurogastroenterology and Motility

mice.47 The pronounced differences between germ-free Behavior has a genetic component, and it is known
mice and mice colonized with complex microbiota that mouse strains differ in their behavioral pheno-
may relate to the ability of gut bacteria to affect type.50,51 There is also a difference in microbiota
multiple aspects of host metabolism, immunity and composition among mouse strains, and the ‘SPF’
physiology. Colonization with a single commensal status does not indicate uniformity of the microbiota,
bacterium, B. thetaiotaomicron was shown to change only the fact that mice have been screened for the
expression of a vast array of genes in the intestine most common murine pathogens. BALB/c and NIH
encoding for metabolism, intestinal permeability, Swiss mice stand on opposite ends of the behavior
angiogenesis but also for glutamate uptake, GABA phenotype: BALB/c mice are timid and less explor-
production and neurotransmitter release.48 atory while NIH Swiss mice display a high explor-
A different approach to investigate the role of atory drive. The BALB/c and NIH Swiss mice were
microbiota in gut-brain axis is to perturb a previously reared under germ-free conditions and then colonized
‘stable’ microbiota in adult healthy mice by oral with SPF microbiota from either NIH Swiss or BALB/c
administration of non-absorbable antimicrobials. mice. Germ-free mice colonized with microbiota from
Combination of neomycin, bacitracin and pimaricin the same strain exhibited similar behavior as the SPF
induced changes in colonic microbiota composition mice. However, mice colonized with microbiota from
(gut dysbiosis) in SPF mice, with a marked increase in the other strain, exhibited a behavior profile similar to
Firmicutes, mainly Lactobacilli spp, and decrease in the donor49 (Fig. 3B). This was not accompanied by any
c-proteobacteria. This was accompanied by an increase measurable change in systemic or gut immune activa-
in mouse exploratory behavior and altered BDNF levels tion or levels of intestinal 5-HT, NA or dopamine. A
in hippocampus and amygdala49 (Fig. 3A). The same change in central neurotrophins was observed one week
antimicrobial treatment failed to induce behavior postcolonization. We can therefore speculate that host
abnormalities in germ-free conditions or in mice behavioral phenotype is also influenced by microbial
treated with antimicrobials intraperitoneally. The factors.
antimicrobial regime employed in this study did not The effect of microbiota may extend into memory
induce measurable changes in gut inflammation or and cognition, as recently suggested in a study
change levels of intestinal serotonin (5-HT), noradren- comparing germ-free and SPF mice.52 An Earlier
alin (NA) or dopamine. Interestingly, studies using study by Li et al. examined the effects of long-term
subdiaphragnatic vagotomy or chemical sympathec- dietary manipulation on memory.53 Mice assigned to
tomy before antimicrobials suggest that vagal and chow with high content of ground beef for 3 months
sympathetic pathways are not involved in gut-brain displayed higher diversity of gut microbiota compared
communication in this experimentally-induced dysbi- to mice on standard rodent chow. This was associated
osis model of altered behavior. with improved working and reference memory, as well

Figure 3 Effects of commensal bacteria on


behavior. (A) Exploratory behavior of adult
BALB/c mice increased transiently in paral-
lel with gut dysbiosis induced by oral anti-
B microbial (ATM) treatment. This was
accompanied by higher brain derived neuro-
trophic factor (BDNF) levels in the hippo-
campus. (B) Colonization of germ-free NIH
Swiss and BALB/c mice with either NIH
Swiss or BALB/c microbiota promoted or
inhibited their exploratory behavior, respec-
tively. Conventional, specific pathogen free
(SPF) mice of both strains were used as
controls. Adapted from Bercik et al.,
Gastroenterology, 2011 (reproduced with
permission from Gastroenterology).

408 Ó 2012 Blackwell Publishing Ltd


Volume 24, Number 5, May 2012 Microbiota-gut-brain axis

as slower speed in seeking food and anxiety-like the enteric nervous system (ENS). The beneficial effect
behavior in mice.53 of probiotic bacteria may extend to healthy individuals.
A study by Desbonnet et al.61 showed that adminis-
Effects of probiotic bacteria on CNS and enteric tration of Bifidobacterium infantis to healthy Sprague-
nervous system function Psychiatric co-morbidities, Dawley rats reduced concentrations of serotonin and
such as anxiety and depression are common in patients dopamine metabolites in the frontal and the amygda-
with chronic bowel disorders, including IBS and loid cortex, respectively. The authors suggested that
inflammatory bowel disease.54–58 Both of these this bacterium might have an anxiolytic potential,
disorders are also associated with abnormal intestinal although no difference in behavior was found in
microbiota profiles.27–30 In this respect, chronic that study. Subsequent experiments with the same
infection with a non-invasive parasite and mild bacterium using maternal separation model demon-
chemically-induced colitis were shown to be associ- strated beneficial effect on altered behavior together
ated with anxiety/depression-like behavior and with normalization of noradrenaline concentrations
decreased levels of hippocampal BDNF expression59,60 in the brainstem.62 Bravo et al. have recently
(Fig. 4). Interestingly, both abnormalities were demonstrated that administration of the probiotic
normalized by the treatment with the probiotic L. rhamnosus JB1 promoted exploratory behavior and
B. longum NC3001 but not with L. rhamnosus attenuated despair-like behavior, as assessed by
NCC4007. Bifidobacterium longum did not improve elevated plus maze and forced swim test, respectively,
gut inflammation or circulating cytokines, however, in healthy BALB/c mice. This was accompanied by a
its anxiolytic effect was absent in mice with previous region-dependent alterations in GABA(B1b) and
vagotomy, suggesting that its action was neurally GABA(Aa2) mRNA in the brain,63 which was vagally
mediated. This was further confirmed by ex vivo dependent, as subdiaphragmatic vagotomy abolished
studies, in which electroresponsiveness of enteric both changes in brain biochemistry and behavior.
neurons was assessed after perfusion with B. longum Thus, animal studies support the notion that
supernatant. Compared to controls, B. longum treated commensal bacteria and specific probiotics can
neurons fired less action potentials in response to influence brain chemistry and the function of the CNS,
supra-threshold depolarizing current.60 The results perhaps by modulating the ENS. This has implications
suggest these signals may be initiated at the levels of for pain perception. Visceral pain perception is regu-

A B

Figure 4 Effects of single probiotic on cen-


tral brain derived neurotrophic factor
(BDNF) and anxiety-like behavior. (A)
Representative micrographs of BDNF
mRNA expression in the hippocampus using
in situ hybridization. BDNF was normalized
in mice with chronic T. muris colitis treated
for 2 weeks with Bifidobacterium longum
(B). (C) Anxiety-like behavior normalized in
mice with T. muris colitis after treatment
with B. longum, but not with Lactobacillus
rhamnosus. Adapted from Bercik et al.,
Gastroenterology, 2011 (reproduced with
permission from Gastroenterology).

Ó 2012 Blackwell Publishing Ltd 409


P. Bercik et al. Neurogastroenterology and Motility

lated by complex mechanisms, including peripheral visceral perception comes from experiments where
sensitization of sensory nerves as well as central microbiota from healthy NIH Swiss mice was delib-
processing, which is modulated by concurrent anxiety erately perturbed by administration of non-absorbable
and depression. Both of these processes could be antibiotics. This treatment resulted in gut dysbiosis
affected by intestinal microbiota. There are multiple characterized by increased levels of Enterobacteriae
clinical trials that have reported improvement of pain and decrease in Lactobacilli and Bacteroides, and low-
after treatment with different probiotic bacteria grade gut inflammation. Mice also developed in-
(Table 1) but the mechanisms of action remain creased visceral perception in response to colorectal
unknown. In one clinical trial, treatment with Bifido- balloon distension accompanied by up-regulation of
bacterium infantis normalized the increased ratio of SP in the myenteric plexus.65 Interestingly, visceral
serum pro-inflammatory cytokines suggesting an hyperalgesia and enteric SP levels normalized after
anti-inflammatory effect may mediate improvement in treatment with Lactobacillus paracasei NCC2461.
pain perception in IBS patients.64 The same probiotic was shown to also reverse rectal
Animal models have provided evidence on possible hyperalgesia in maternally deprived rats.66 The effect
neural and metabolic pathways affected by commensals of probiotics on pain mechanisms extends beyond an
and probiotics. The first report on the role of bacteria on effect on enteric nerves as Lactobacillus farciminis
was shown to ameliorate stress-induced visceral
hyperalgesia and affect neural activation patterns
Table 1 Randomized control trials showing beneficial effect of assessed by c-fos staining in the sacral spine, parav-
probiotics on abdominal pain
entricular nucleus of hypothalamus and medial amyg-
daloid nucleus.67
Reference Probiotic treatment
Several probiotic bacteria have been tested for their
Niedzielin et al., Eur J Gastroenterol Lactobacillus plantarum ability to decrease pain perception during colorectal
Hepatol 2001 299V
distension. Bifidobacterium infantis 35624 was shown
O’Mahony et al., Gastroenterology Bifidobacterium infantis
2005 35624 to be effective in reducing visceral pain in both visceral
Bausserman et al., J Pediatr 2005 Lactobacillus GG normosensitive (Sprague-Dawley) and visceral hyper-
Whorwell et al., J Gastroenterol 2006Bifidobacterium infantis
sensitive (Wistar-Kyoto) rats.68 Lactobacillus reuteri is
35624
Gawronska et al., Alim Pharmacol Lactobacillus GG a well- characterized strain with respect to its antin-
Ther 2007 ociceptive effect: its administration decreased response
Sinn et al., Dig Dis Sci 2008 Lactobacillus acidophilus
to colorectal distension in rats69 and prevented hype-
SDC 2012
Drouault-Holowacz et al., B. longum LA101, rexcitability of colonic DRG neurons induced by
Gastroenterol Clin Biol 2008 L. acidophilus LA102, noxious stimuli.70 A subsequent study showed that
L. lactis LA103, S. thermophilus
L. reuteri increases excitability and number of action
LA104
Enck et al., Neurogastroeterol E. coli DSM 17252, potentials per depolarizing pulse, decreases calcium-
Motility 2008 Enterococcus faecalis DSM dependent potassium channel opening and decreases
16440
the slow after hyperpolarization in sensory AH neu-
Andriulli et al., J Clin Gastroenterol L. paracasei B 21060,
2008 L. paracasei B21070, rons.71 It is important to note that the effect of a given
L. gasseri B21090 probiotic on visceral pain is likely site specific, as a
Williams et al., Alim Pharmacol L. acidophilus CUL60,
recent study has shown that L. reuteri but not Lacto-
Ther 2009 L. acidophilus CUL21,
B. lactis CUL34, B. bifidum bacillus plantarum alleviates the response to gastric
CUL20 distension in rats, although both of them have previ-
Enck et al., Z Gastroenterol 2009 E.coli DSM17252
ously been shown to be effective anti-nociceptive
Kalman et al., BMC Gastroenterol Bacillus coagulans GBI-30
2009 agents in colorectal distension.72
Guandalini et al., J Pediatr VSL#3 preparation The mechanisms of action of specific probiotics on
Gastroenterol Nutr 2010
visceral pain perception and ENS modulation are likely
Romano et al., J Paediatr Child L. reuteri DSM 17938
Health 2010 to be multiple and strain specific. The D-alanine
Francavilla et al., Pediatrics 2010 Lactobacillus GG content of lipoteichoic acid was found to be crucial
Guglielmetti et al., Aliment B. bifidum MIMBb75
for Lactobacillus plantarum-mediated protection from
Pharmacol Ther 2011
Guerra et al., World J Gastroenterol B. longum (clinical isolate) visceral pain perception in rats.73 Another probiotic,
2011 Lactobacillus acidophilus NCFM induced the expres-
Tarrerias et al., Dig Dis 2011 Lactobacillus LB
sion of opioid and cannabinoid receptors in intestinal
VSL#3Ò: B. breve, B. longum, B. infantis, L. acidophilus, L.plantarum, epithelial cells, and modulated analgesic functions in
L. paracasei, L. bulgaricus, S. thermophilus. the gut of healthy rats.74

410 Ó 2012 Blackwell Publishing Ltd


Volume 24, Number 5, May 2012 Microbiota-gut-brain axis

translate into our ability to diagnose and treat patients


Challenges of studying the role of microbiota
with both gut and brain disorders.
in the gut-brain axis
Although here is accumulating evidence for bacteria
CONCLUSIONS
playing an important role in gut-brain axis, our
knowledge is limited by our ability to identity the While clinical observation and psychiatric co-morbid-
key players in this complex bacterial community. ity in various chronic intestinal disorders support a role
Most of the evidence comes from animal experiments of the intestinal microbiota in gut-brain axis commu-
where the microbiota is less diverse than in humans nication, the strongest evidence for a role of microbes
and can be controlled experimentally (e.g. gnotobiotic as signaling components in the gut-brain axis comes
conditions). In the last decade, significant advance- form from animal studies using perturbation of the
ment has been provided by the availabiltity of molec- microbiota by antimicrobials and gnotobiotic models.
ular techniques, such as deep sequencing. These Mechanisms of communication are likely to be multi-
techniques enable us to determine bacteria at the ple and involve neural, humoral and inflammatory
species level. However, the task to identify a microor- pathways, depending on host and environmental fac-
ganism responsible for effects on the CNS is challeng- tors. Animal studies will be crucial to continue to
ing. First, the key bacterium could be underrepresented provide mechanistic insight and proof-of concept
in the community and could be missed when scanning studies. However, there is need for the concepts
for total microbiota composition. Also, results are generated in animal models to be translated to the
influenced by the technique employed in the analysis human in the future.
of microbiota (454 pyrosequencing, Illumina platform,
microarrays). The choice of technique can also make
ACKNOWLEDGMENT
the comparison between individual studies difficult.
Second, even if we identify the key microorganism(s) it We thank Dr Neil Dufton (http://www.notproperlydone.com) for
artistic design of Figs 1 and 2. PB is a recipient of HHS Early
may be difficult to culture and to determine its
Career Award.
characteristics. Last, it is possible that the action of
microbiota on the CNS is not related to a specific
bacterium or group of bacteria, but rather to their FUNDING
metabolic activity. This metabolic profile may be The authors are partially funded by CIHR, CCFC, CCA, and a
influenced by environmental factors such as diet, grant from Nestle Research Center.
stress or inflammation. Thus, a metabolomic approach
would be more likely to yield answers than identifica-
DISCLOSURES
tion of single bacteria. Despite these methodological
set-backs, our understanding of microbial-host inter- SMC is on Advisory Board of Salix and Institute Rosell.
actions is rapidly advancing and this will likely

REFERENCES 5 Hooper LV, Macpherson AJ. Res Clin Gastroenterol 2004; 18: 315–
Immune adaptations that maintain 21.
1 Banks WA. Evidence for a cholecys- homeostasis with the intestinal 9 Hooper LV, Gordon JI. Commensal
tokinin gut-brain axis with modula- microbiota. Nat Rev Immunol 2010; host-bacterial relationships in the
tion by bombesin. Peptides 1980; 1: 10: 159–69. gut. Science 2001; 292: 1115–8.
347–51. 6 Bäckhed F, Ley RE, Sonnenburg JL 10 Dumas ME, Barton RH, Toye A et al.
2 Mayer EA. Gut feelings: the emerging et al. Host-bacterial mutualism in Metabolic profiling reveals a contri-
biology of gut-brain communication. the human intestine. Science 2005; bution of gut microbiota to fatty
Nat Rev Neurosci 2011; 12: 453–66. 307: 1915–20. liver phenotype in resistant mice. Proc
3 O’Mahony SM, Hyland NP, Dinan 7 Husebye E, Hellstrom PM, Sundler F Natl Acad Sci USA 2006; 103:
TG, Cryan JF. Maternal separation as et al. Influence of microbial species 12511–6.
a model of brain-gut axis dysfunction. on small intestinal myoelectric 11 Zoetendal E, Akkermans AD, DeVos
Psychopharmacology 2011; 214: 71– activity and transit in germ-free rats. WM. Temperature gradient gel elec-
88. Am J Physiol Gastrointest Liver trophoresis analysis of 16S rRNA
4 Eckburg PB, Bik EM, Bernstein CN Physiol 2001; 280: G368–80. from human fecal samples reveals
et al. Diversity of the human intesti- 8 Verdu EF, Collins SM. Microbial-gut stable and host-specific communities
nal microbial flora. Science 2005; 308: interactions in health and disease. of active bacteria. Appl Environ
1635–8. Irritable bowel syndrome. Best Pract Microbiol 1998; 64: 3854–9.

Ó 2012 Blackwell Publishing Ltd 411


P. Bercik et al. Neurogastroenterology and Motility

12 Gronlund MM, Lehtonen OP, Eerola 23 McBurney WT, McCartney A, Apun 34 Posey DJ, Kem DL, Swiezy NB et al.
E, Kero P. Fecal microflora in healthy A et al. Perturbation of the entero- A pilot study of D-cycloserine in
infants born by different methods of bacterial microflora detected by subjects with autistic disorder. Am J
delivery: permanent changes in molecular analysis. Microb Ecol Psychiatry 2004; 161: 2115–7.
intestinal flora after cesarean deliv- Health Dis 1999; 11: 75–9. 35 Mehdi S. Antibiotic-induced psycho-
ery. J Pediatr Gastroenterol Nutr 24 Löfmark S, Jernberg C, Jansson JK, sis: a link to D-alanine? Med
1999; 28: 19–25. Edlund C. Clindamycin-induced Hypotheses 2010; 75: 676–7.
13 Martin R, Heilig GH, Zoetendal EG enrichment and long-term persis- 36 Ledochowski M, Sperner-Unterweger
et al. Diversity of the Lactobacillus tence of resistant Bacteroides spp. B, Fuchs D. Lactose malabsorption is
group in breast milk and vagina of and resistance genes. J Antimicrob associated with early signs of mental
healthy women and potential role in Chemother 2006; 58: 1160–7. depression in females: a preliminary
the colonization of the infant 25 Jernberg C, Löfmark S, Edlund C, report. Dig Dis Sci 1998; 43: 2513–7.
gut. J Appl Microbiol 2007; 103: Jansson JK. Long-term ecological 37 Ledochowski M, Widner B, Sperner-
2638–44. impacts of antibiotic administration Unterweger B et al. Carbohydrate
14 Coppa GV, Bruni S, Morelli L et al. on the human intestinal microbiota. malabsorption syndromes and early
The first prebiotics in humans: ISME J 2007; 1: 56–66. signs of mental depression in females.
human milk oligosaccharides. J Clin 26 De FilippoC, Cavalieri D, Di Paola M Dig Dis Sci 2000; 45: 1255–9.
Gastroenterol 2004; 38: S80–3. et al. Impact of diet in shaping gut 38 Lyte M, Varcoe JJ, Bailey MT. Anxi-
15 Lewis S, Cochrane S. Alteration of microbiota revealed by a comparative ogenic effect of subclinical bacterial
sulfate and hydrogen metabolism in study in children from Europe and infection in mice in the absence of
the human colon by changing intes- rural Africa. Proc Natl Acad Sci USA overt immune activation. Physiol
tinal transit rate. Am J Gastroenterol 2010; 107: 14691–6. Behav 1998; 65: 63–8.
2007; 102: 624–33. 27 Maukonen J, Satokari R, Mättö J et al. 39 Gaykema RP, Goehler LE, Lyte M.
16 Tannock GW, Munro K, Harmsen Prevalence and temporal stability of Brain response to cecal infection
HJM et al. Analysis of the fecal selected clostridial groups in irritable with Campylobacter jejuni: analysis
microflora of human subjects con- bowel syndrome in relation to pre- with Fos immunohistochemistry.
suming a probiotic containing Lacto- dominant faecal bacteria. J Med Brain Behav Immun 2004; 18: 238–
bacillus rhamnosus DR20. Appl Microbiol 2006; 55: 625–33. 45.
Environ Microbiol 2000; 66: 2578–88. 28 Mättö J, Maunuksela L, Kajander K 40 Goehler LE, Park SM, Opitz N et al.
17 Zoetendal EG, Akkermans ADL, Vli- et al. Composition and temporal sta- Campylobacter jejuni infection
et WMA et al. The Host Genotype bility of gastrointestinal microbiota increases anxiety-like behavior in the
Affects the Bacterial Community in in irritable bowel syndrome – a lon- holeboard: possible anatomical sub-
the Human Gastronintestinal Tract. gitudinal study in IBS and control strates for viscerosensory modulation
Microb Ecol Health Dis 2001; 13: subjects. FEMS Immunol Med of exploratory behavior. Brain Behav
129–34. Microbiol 2005; 43: 213–22. Immun 2008; 22: 354–66.
18 Arumugam M, Raes J, Pelletier E et al. 29 Martinez C, Antolin M, Santos J et al. 41 Lyte M, Li W, Opitz N et al. Induc-
Enterotypes of the human gut mi- Unstable composition of the fecal tion of anxiety-like behavior in mice
crobiome. Nature 2011; 473: 174–80. microbiota in ulcerative colitis dur- during the initial stages of infection
19 Wu GD, Chen J, Hoffmann C et al. ing clinical remission. Am J Gastro- with the agent of murine colonic
Linking long-term dietary patterns enterol 2008; 103: 643–8. hyperplasia Citrobacter rodentium.
with gut microbial enterotypes. Sci- 30 Scanlan PD, Shanahan F, O’Mahony Physiol Behav 2006; 89: 350–7.
ence 2011; 334: 105–8. C, Marchesi JR. Culture-independent 42 Goehler LE, Gaykema RP, Opitz N
20 McCartney AL, Wenzhi W, Tannock analyses of temporal variation of the et al. Activation in vagal afferents
GW. Molecular analysis of the com- dominant fecal microbiota and tar- and central autonomic pathways:
position of the bifidobacterial and geted bacterial subgroups in Crohn’s early responses to intestinal infection
lactobacillus microflora of humans. disease. J Clin Microbiol 2006; 44: with Campylobacter jejuni. Brain
Appl Environ Microbiol 1996; 62: 3980–8. Behav Immun 2005; 19: 334–44.
4608–13. 31 Bass NM. Review article: the current 43 Bercı́k P, De GiorgioR, Blennerhas-
21 Bartosch S, Fite A, Macfarlane GT, pharmacological therapies for hepatic sett P et al. Immune-mediated neural
McMurdo ME. Characterization of encephalopathy. Aliment Pharmacol dysfunction in a murine model of
bacterial communities in feces from Ther 2007; 25(Suppl 1): 23–31. chronic Helicobacter pylori infection.
healthy elderly volunteers and hospi- 32 Parracho HM, Bingham MO, Gibson Gastroenterology 2002; 123: 1205–15.
talized elderly patients by using real- GR, McCartney AL. Differences 44 Bercik P, Verdú EF, Foster JA et al.
time PCR and effects of antibiotic between the gut microflora of chil- Role of gut-brain axis in persistent
treatment on the fecal microbiota. dren with autistic spectrum disorders abnormal feeding behavior in mice
Appl Environ Microbiol 2004; 70: and that of healthy children. J Med following eradication of Helicobacter
3575–81. Microbiol 2005; 54: 987–91. pylori infection. Am J Physiol Regul
22 Kleessen B, Schroedl W, Stueck M 33 Sandler RH, Finegold SM, Bolte ER Integr Comp Physiol 2009; 296:
et al. Microbial and immunological et al. Short-term benefit from oral R587–94.
responses relative to high-altitude vancomycin treatment of regressive- 45 Sudo N, Chida Y, Aiba Y et al. Post-
exposure in mountaineers. Med Sci onset autism. J Child Neurol 2000; natal microbial colonization programs
Sports Exerc 2005; 37: 1313–8. 15: 429–35. the hypothalamic-pituitary-adrenal

412 Ó 2012 Blackwell Publishing Ltd


Volume 24, Number 5, May 2012 Microbiota-gut-brain axis

system for stress response in mice. Arch Psychiatry Clin Neurosci 2003; 66 Eutamene H, Lamine F, Chabo C
J Physiol 2004; 558: 263–75. 253: 313–20. et al. Synergy between Lactobacillus
46 Heijtz RD, Wang S, Anuar F et al. 57 Fuller-Thomson E, Sulman J. Depres- paracasei and its bacterial products to
Normal gut microbiota modulates sion and inflammatory bowel disease: counteract stress-induced gut perme-
brain development and behavior. Proc findings from two nationally repre- ability and sensitivity increase in
Natl Acad Sci U S A 2011; 108: 3047– sentative Canadian surveys. Inflamm rats. J Nutr 2007; 137: 1901–7.
52. Bowel Dis 2006; 12: 697–707. 67 Ait-Belgnaoui A, Eutamene H, Hou-
47 Neufeld KM, Kang N, Bienenstock J, 58 Walker JR, Ediger JP, Graff LA et al. deau E et al. Lactobacillus farciminis
Foster JA. Reduced anxiety-like The Manitoba IBD cohort study: a treatment attenuates stress-induced
behavior and central neurochemical population-based study of the preva- overexpression of Fos protein in
change in germ-free mice. Neurogas- lence of lifetime and 12-month anxi- spinal and supraspinal sites after
troenterol Motil 2011; 23: 255–64. ety and mood disorders. Am J colorectal distension in rats. Neuro-
48 Hooper LV, Wong MH, Thelin A et al. Gastroenterol 2008; 103: 1989–97. gastroenterol Motil 2009; 21: 567–73.
Molecular analysis of commensal 59 Bercik P, Verdu EF, Foster JA et al. 68 McKernan DP, Fitzgerald P, Dinan
host-microbial relationships in the Chronic gastrointestinal inflamma- TG, Cryan JF. The probiotic Bifido-
intestine. Science 2001; 291: 881–4. tion induces anxiety-like behavior bacterium infantis 35624 displays
49 Bercik P, Denou E, Collins J et al. and alters central nervous system visceral antinociceptive effects in the
The intestinal microbiota affect cen- biochemistry in mice. Gastroenterol- rat. Neurogastroenterol Motil 2010;
tral levels of brain-derived neuro- ogy 2010; 139: 2102–12. e1. 22: 1029–35.
tropic factor and behavior in mice. 60 Bercik P, Park AJ, Sinclair D et al. 69 Kamiya T, Wang L, Forsythe P et al.
Gastroenterology 2011; 141: 599–609. The anxiolytic effect of Bifidobacte- Inhibitory effects of Lactobacillus
50 Conti LH, Costello DG, Martin LA rium longum NCC3001 involves reuteri on visceral pain induced by
et al. Mouse strain differences in the vagal pathways for gut-brain com- colorectal distension in Sprague-
behavioral effects of corticotropin- munication. Neurogastroenterol Dawley rats. Gut 2006; 55: 191–6.
releasing factor (CRF) and the CRF Motil 2011; 23: 1132–9. 70 Ma X, Mao YK, Wang B et al. Lacto-
antagonist alpha-helical CRF9-41. 61 Desbonnet L, Garrett L, Clarke G, bacillus reuteri ingestion prevents
Pharmacol Biochem Behav 1994; 48: Bienenstock J, Dinan TG. The probi- hyperexcitability of colonic DRG
497–503. otic Bifidobacteria infantis: an neurons induced by noxious stimuli.
51 Kalinichev M, Bate ST, Coggon SA assessment of potential antidepres- Am J Physiol Gastrointest Liver
et al. Locomotor reactivity to a novel sant properties in the rat. J Psychiatr Physiol 2009; 296: G868–75.
environment and sensitivity to Res 2008; 43: 164–74. 71 Kunze WA, Mao YK, Wang B et al.
MK-801 in five strains of mice. Behav 62 Desbonnet L, Garrett L, Clarke G Lactobacillus reuteri enhances excit-
Pharmacol 2008; 19: 71–5. et al. Effects of the probiotic Bifido- ability of colonic AH neurons by
52 Gareau MG, Wine E, Rodrigues DM bacterium infantis in the maternal inhibiting calcium-dependent potas-
et al. Bacterial infection causes separation model of depression. Neu- sium channel opening. J Cell Mol
stress-induced memory dysfunction roscience 2010; 170: 1179–88. Med 2009; 13: 2261–70.
in mice. Gut 2011; 60: 307–17. 63 Bravo JA, Forsythe P, Chew MV 72 Duncker SC, Kamiya T, Wang L et al.
53 Li W, Dowd SE, Scurlock B et al. et al. Ingestion of Lactobacillus Probiotic Lactobacillus reuteri
Memory and learning behavior in strain regulates emotional behavior alleviates the response to gastric
mice is temporally associated with and central GABA receptor expres- distension in rats. J Nutr 2011; 141:
diet-induced alterations in gut bacte- sion in a mouse via the vagus nerve. 1813–8.
ria. Physiol Behav 2009; 96: 557–67. Proc Natl Acad Sci U S A 2011; 73 Duncker SC, Wang L, Hols P, Bie-
54 Folks DG. The interface of psychiatry 108: 16050–5. nenstock J. The D-alanine content of
and irritable bowel syndrome. Curr 64 O’Mahony L, McCarthy J, Kelly P lipoteichoic acid is crucial for Lacto-
Psychiatry Rep 2004; 6: 210–5. et al. Lactobacillus and Bifidobacte- bacillus plantarum-mediated protec-
55 Whitehead WE, Palsson O, Jones KR. rium in irritable bowel syndrome: tion from visceral pain perception in a
Systematic review of the comorbidity symptom responses and relationship rat colorectal distension model. Neu-
of irritable bowel syndrome with to cytokine profiles. Gastroenterol- rogastroenterol Motil 2008; 20: 843–
other disorders: what are the causes ogy 2005; 128: 541–51. 50.
and implications? Gastroenterology 65 Verdú EF, Bercik P, Verma-Gandhu M 74 Rousseaux C, Thuru X, Gelot A et al.
2002; 122: 1140–56. et al. Specific probiotic therapy Lactobacillus acidophilus modulates
56 Harter MC, Conway KP, Merikangas attenuates antibiotic induced visceral intestinal pain and induces opioid and
KR. Associations between anxiety hypersensitivity in mice. Gut 2006; cannabinoid receptors. Nat Med
disorders and physical illness. Eur 55: 182–90. 2007; 13: 35–7.

Ó 2012 Blackwell Publishing Ltd 413

You might also like