El Eje Microbiota Intestinal - Cerebro y La Salud Mental (Dinan y Cryan, 2017)

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

Psychosomatic Medicine

Author’s Accepted Manuscript

Article Title: Brain-gut-microbiota axis in psychiatry:


novel paradigm or false dawn?

Authors: Timothy G. Dinan and John Cryan

D
DOI: 10.1097/PSY.0000000000000519

Received Date: July 29, 2016


Revised Date: Apr 27, 2017

TE
EP
This manuscript has been accepted by the editors of Psychosomatic Medicine, but it
has not yet been copy edited; information within these pages is therefore subject to
change. During the copy-editing and production phases, language usage and any
textual errors will be corrected, and pages will be composed into their final format.

Please visit the journal’s website (www.psychosomaticmedicine.org) to check for a


C

final version of the article.

When citing this article, please use the following: Psychosomatic Medicine (in press)
and include the article’s digital object identifier (DOI).
C
A

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Psychosomatic Medicine Publish Ahead of Print
DOI: 10.1097/PSY.0000000000000519

Brain-gut-microbiota axis and mental health

Timothy G. Dinan, MD, PhD1,2, and John F. Cryan, PhD1,3

1
APC Microbiome Institute, University College Cork, Cork, Ireland

D
2
Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork,

TE
Ireland

3
Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
EP
Corresponding author:

Prof. Ted Dinan, Department of Psychiatry, University College Cork, Ireland. t.dinan@ucc.ie
C

Conflicts of interest and Source of Funding


C

No conflicts of interest to declare. The authors are supported in part by Science Foundation

Ireland in the form of a centre grant (Alimentary Pharmabiotic Centre Grant Number
A

SFI/12/RC/2273); by the Health Research Board of Ireland (Grant Numbers

HRA_POR/2011/23 and HRA_POR/2012/32) and received funding from the European

Community’s Seventh Framework Programme Grant MyNewGut under Grant Agreement

No. FP7/2007-2013. The Centre has conducted studies in collaboration with several

companies including GSK, Pfizer, Cremo, Suntory, Wyeth and Mead Johnson.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Abstract

OBJECTIVE: The brain-gut-microbiota axis has been put forward as a new paradigm in

neuroscience, which may be of relevance to mental illness. The mechanisms of signal

transmission in the brain-gut-microbiota axis are complex and involve bidirectional

communications which enables gut microbes to communicate with the brain, and the brain to

communicate with the microbes. This review assesses the potential usefulness and limitations

of the paradigm.

D
METHODS: A selective literature review was conducted to evaluate the current knowledge

TE
in clinical and pre-clinical brain-gut-microbiota interactions as related to psychiatric

disorders.

RESULTS: The overwhelming majority of published studies in the field are preclinical and
EP
there is so far a lack of clinical studies. Preliminary studies in psychiatric populations,

support the view of a dysbiosis in some conditions, but studies are often small-scale and

marred by potential confounding variables. Preclinical studies support the view that

psychobiotics (‘bacteria which when ingested in adequate amounts have a positive mental
C

health benefit’) might be of use in treating some patients with mental health difficulties. To

date we have no well conducted studies in clinical populations, though there are some studies
C

in healthy volunteers. A cocktail of probiotics has been shown to alter brain activity as

monitored by functional MRI and Bifidobacterium longum was reported to alter brain
A

electrical activity.

CONCLUSION: It has yet to be convincingly demonstrated that the exciting findings of

psychobiotic efficacy demonstrated in preclinical models of psychiatric illness will translate

to patients.

Key words: Brain-gut-microbiota axis, psychobiotics, depression, autism, anxiety,

schizophrenia, bifidobacteria, lactobacilli

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Abbreviations

BBB Blood brain barrier

CBT cognitive behavior therapy

C. difficile Clostridium difficile

CRF corticotropin releasing factor

D
cFos immediate early gene product

5HT Serotonin

FOS

GABA TE
Fructooligosaccharides

Gamma-aminobutyric acid
EP
GOS Galactooligosaccharides

GPCR G protein coupled receptor

HDACs histone deacetylases


C

HPA Hypothalamic-pituitary-adrenal axis


C

IBS Irritable bowels syndrome


A

IL Interleukin

L. rhamnosus Lactobacillus rhamnosus

SCFAs Short chain fatty acids

SSRIs Selective serotonin reuptake inhibitors

WAS water avoidance stress

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
The complexity of psychiatric illnesses cannot be underestimated and despite major advances

in molecular neurobiology and neuroimaging, there have been no major advances in

therapeutics. As a discipline psychiatry has had many false dawns and the objective of this

review is to assess the newly emerging paradigm of the brain-gut-microbiota axis and

critically assess its value in generating new therapeutic strategies.

The nineteen fifties were a watershed in the history of psychiatry with the emergence of the

D
first effective antipsychotics and antidepressants [1]. The demonstrated efficacy of

chlorpromazine in treating schizophrenia, a molecule synthesized by Charpentier, was

TE
undoubtedly a landmark [2]. In terms of depression treatment, the antidepressant iproniazid

emerged [3]. It is a monoamine-oxidase inhibitor that had originally been used in the

treatment of tuberculosis. Shortly afterwards imipramine, the first tricyclic antidepressant

was synthesised and originally considered useful as an antipsychotic. However, it soon


EP
became clear it has little efficacy in this regard but showed promise in the management of

depression [4].

Since the nineteen fifties there have been no fundamental new advances in
C

psychopharmacology. Undoubtedly, the emergence of SSRIs such as fluoxetine in the

eighties was an advance in the treatment of depression in terms of side-effects. However, the
C

SSRIs failed to yield improvements in terms of efficacy or speed of therapeutic onset [5].

Likewise, the emergence of atypical antipsychotics such as olanzapine and risperidone


A

provided treatments without the extrapyramidal side-effects of the earlier compounds, but

they are no more efficacious (with the exception of clozapine) and prone to cause

considerable weight gain frequently leading to a metabolic syndrome [6].

There are those who would argue that the major new paradigm in psychiatry has been the

emergence of more effective psychological interventions such as cognitive behaviour therapy

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
(CBT) and mindfulness. Hundreds of studies have been published on mindfulness in the past

few years and almost all have reported positive findings[7]. While these therapies benefit

many patients their efficacy may be exaggerated by the large number of studies with small

samples sizes and very varying definitions of response [8]. Many studies of these therapies

have used designs which exaggerate the effect size of the interventions [9, 10]. Few if any of

the studies are designed with the capacity to compare efficacy with a true placebo response.

Many for example use a waiting list comparison or a comparison with treatment as usual.

D
Both of these controls will exaggerate the effect size of any intervention. A recent meta-

analysis of CBT trials concludes that control conditions are often less than optimal [11].

appropriately.

Evidence for novel paradigm


TE
Despite such reservations, many patients do benefit from these treatments when used
EP
The brain-gut-microbiota axis is the focus of the latest paradigm in neuroscience that has

been put forward as a potential game changer [12, 13]. An exponentially accumulating

volume of evidence supports the view that gut microbes have a profound impact on central
C

neurochemistry and behaviour, especially stress related responses[14]. How do gut microbes

exert such a powerful central influence and how might targeting the brain-gut-microbiota axis
C

yield effective therapies for psychiatric illnesses?


A

Until February 2017, there were 142 articles focusing on gut microbes and the brain, listed on

PubMed. Twenty-three of these articles centred on gut microbes and mental illness, while 35

papers focused on psychobiotics. Taking a broader perspective, looking at probiotics and

mental health there were 50 papers. Of the 142 papers on the gut microbiota and the brain

111 were reviews and 31 were experimental studies of which only 4 were human studies.

This latter fact pinpoints the current weakness of this nascent field.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Microbe to brain communication

The brain-gut-microbiota axis (Figure 1) is a bidirectional communication system which

enables gut microbes to communicate with the brain, and the brain in turn to communicate

with the gut [15]. While brain-gut communication has been a subject of investigation for

decades an exploration of gut microbes as a mediator within this context has only recently

been addressed. The mechanisms of signal transmission are complex and not fully elucidated,

D
but it is clear they include neural, endocrine, immune, and metabolic pathways [16-18].

Preclinical studies have implicated the vagus nerve as a fundamental neural route of

TE
communication between gut microbes in the periphery and centrally-mediated behavioral

effects, as illustrated by the elimination of central Lactobacillus rhamnosus effects following

full truncal vagotomy [19]. Interestingly, it has been demonstrated that individuals who
EP
underwent a full truncal vagotomy for treatment of peptic ulcer disease have a decreased risk

of certain neurological disorders such as Parkinson’s disease when they enter old age [20].

The gut microbiota also regulates central neurotransmitters such as serotonin by altering

levels of precursors; for example Bifidobacterium infantis has been demonstrated to elevate
C

plasma tryptophan levels and thus influence central 5-HT [21]. Tryptophan is the precursor
C

of 5HT and the human brain has limited storage capacity, therefore requiring a continual

replenishment from the intestine. As well as producing precursors many bacteria can
A

synthesise and release neurotransmitters e.g. Lactobacillus and Bifidobacterium species can

produce gamma-aminobutyric acid (GABA): Escheridia, Bacillus and Saccharomyces spp.

can produce norepinephrine: Candida, Streptococcus, Escheridia and Enterococcus spp. can

produce 5HT: Bacillus can produce dopamine: and Lactobacillus can produce acetylcholine

[22, 23]. These microbe-produced neurotransmitters can cross the mucosal layer of the

intestine, though it is improbable that they directly influence brain physiology. Even if they

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
enter the blood stream, which is by no means certain, they are incapable of crossing the blood

brain barrier (BBB). Their impact on brain function is likely to occur by acting locally on the

enteric nervous system. Short chain fatty acids (SCFAs), which include butyrate, propionate

and acetate are essential metabolic products of gut microbial activity and may exert central

effects either through GPCRs (G-protein coupled receptors), though such receptors are

sparsely concentrated in the mammalian brain and the half-life of SCFAs is exceedingly short

in the plasma. They may however act as epigenetic modulators through histone deacetylases

D
(HDACs)[24]. Immune signalling from gut to brain mediated by cytokine molecules is

TE
another well documented route of communication [25]. Cytokines produced at the level of the

gut can travel via the bloodstream to the brain. Under normal physiological circumstances

they do not cross the BBB, but increasing evidence indicates an ability to signal across the

BBB and to influence brain areas such as the hypothalamus and circumventricular organs
EP
where the BBB is deficient. It is through the latter mechanism the cytokines interleukin (IL)-

1 and IL-6 activate the core stress system, the hypothalamic-pituitary-adrenal axis (HPA),

bringing about the release of cortisol. This is regarded as the most potent activating
C

mechanism of the stress system and is of relevance in conditions such as the depression that

emerges with interferon therapy for hepatitis or induced by infections [26] and also
C

inflammatory bowel disorders and irritable bowel syndrome which frequently have

psychiatric co-morbidities[27].
A

Psychopathology and gut dysbiosis

There is increasing evidence that some psychiatric and neurological disorders may be

associated with a gut dysbiosis. The extent to which such a dysbiosis is central to the

pathophysiology of these conditions has not been fully elucidated.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Depression

Dinan and his colleagues studied the gut microbiota in a maternal separation model of

depression in rats [28]. They reported an overactive HPA response in such animals, together

with an increase in pro-inflammatory cytokines and a decrease in the diversity of gut

microbes. In a recent study the fecal microbiota was sequenced in a depression study [29].

Forty-six patients with depression and 30 healthy controls were recruited. . High-throughput

D
pyrosequencing showed that, according to the Shannon index, increased fecal bacterial alpha-

diversity was found in those currently depressed but not in a group who had responded to

TE
treatment. Bacteroidetes, Proteobacteria, and Actinobacteria were increased, whereas

Firmicutes were significantly reduced. Despite the profound inter-individual variability,

levels of several predominant genera were differed between the depressives and controls.

Most notably, the depressives had increased levels of Enterobacteriaceae and Alistipes but
EP
reduced levels of Faecalibacterium. The authors conclude that further studies are necessary to

elucidate the temporal and causal relationships between gut microbiota and depression and to

evaluate the suitability of the microbiome as a biomarker. In our study depressed patients
C

had elevated cortisol output together with decreased faecal microbial richness and when rats

were given a humanised microbiota from depressed patients, as opposed to healthy controls,
C

they developed a depressive phenotype from a behavioural and immune perspective[30].

Anxiety disorders
A

There are no published studies exploring the gut microbiota in any specific anxiety disorder.

Of the anxiety disorders, obsessive compulsive disorder (OCD) has been most consistently

associated with infection, especially respiratory tract infection with group A beta-hemolytic

streptoccus [31]. No studies of probiotics in OCD patients have been undertaken, though a

rodent study suggests that L. rhamnosus might be effective [32].

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Lyte et al [33] found that oral gavage of the pathogen Campylobacter jejuni, in subclinical

doses, which did not produce an overt immune response, resulted in anxiety-type behaviour

in rodents. They also noted that areas of brainstem, such as the nucleus tractus solitarius and

lateral parabrachial nucleus, are involved in the processing that results in autonomic,

neuroendocrine and behavioural responses.

In a recently published study Bruch interrogated the Medical Expenditure Panel Survey

D
(MEPS) to prospectively determine the relationship between intestinal infection and future

onset of an anxiety disorder [34]. A total of six 2-year panel datasets, which consisted of 5

TE
consecutive rounds, were amalgamated from 2007 to 2013. This included the data for all

respondents who were 18 years of age or older and who importantly did not have an anxiety

disorder at baseline. Within the study population, there were 2577 subjects with an intestinal
EP
infection in Round 1 and 4239 with an anxiety disorder that commenced in Round 2, 3, 4, or

5. In total an intestinal infection in Round 1 was associated with a 1.34 (P < 0.01) odds ratio

of an emerging anxiety disorder starting in Round 2, 3, 4, or 5. Does this association apply


C

to other infections such as respiratory or urinary tract? Respiratory infection was associated

with a 1.36 (P < 0.01) odds ratio of having an anxiety disorder that began in Round 2, 3, 4, or
C

5 but other infections were unrelated. This major epidemiological study provides convincing
A

evidence of a link between intestinal infection and the subsequent development of anxiety.

Autism

Autism is a neurodevelopmental disorder whose prevalence is regarded by some as on the

increase[35]. It is characterised by a failure of language acquisition, obsessional traits and a

lack of sociability and is frequently associated with gastrointestinal symptoms [36], the

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
relevance of which has been a longstanding source of controversy. Up to 70% of patients

with the syndrome report abdominal symptoms and hence the view that it is a disorder of the

brain-gut axis[37].

At the APC Microbiome Institute the behaviour of mice raised in a germ-free environment

was examined [38, 39]. The mice were tested in a three chamber apparatus which assesses

social interaction. Here, a germ-free animal was put in the middle chamber with a familiar

D
mouse in chamber 1 and a novel mouse in chamber 3. The germ-free mouse interacted with

the familiar mouse for a similar amount of time to the novel mouse. This contrasts with the

TE
behaviour of conventionally colonised mice, who spend less time with the familiar than the

novel mouse. Germ-free mice also interact with objects to the same exact as with other

animals, a very unusual pattern of behaviour for a sociable animal. Conventional


EP
colonisation of the germ free mice does to some extent normalise their behaviour. The

behavioural anomalies s are related to underlying alterations in neural circuitry.

Work from Mazmanian’s group in an animal model demonstrated that the microbiota

mediates the behavioural and biochemical anomalies associated with neurodevelopmental


C

disorders including autism [40]. They made use of the maternal immune activation model
C

which they induced by poly-IC injection during pregnancy and found significant changes in

gastrointestinal barrier function as well as microbiota anomalies. Oral treatment with the
A

human commensal Bacteroides fragilis was shown to normalise gut permeability and most

interestingly stereotypic and other aberrant behaviours. Even more importantly, a metabolite

found in the abnormal animals was observed to transfer the phenotype to naïve animals and

to be reduced by Bacteroides fragilis.

There is currently a lot of attention focused on oxytocin, the hypothalamic peptide, released

from the posterior pituitary, which is has been demonstrated to increase sociability. The

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
oxytocin receptor knockout mouse in behavioural tests has major deficits in social behaviour

[41] and small scale studies in humans have shown that that intra-nasally administered

oxytocin positively impacts social behaviour. Large scale clinical trials of oxytocin in autism

spectrum disorder are currently under way [42]. There is controversy as to whether or not the

preclinical findings translate to the clinical setting and assuming they do which subset of

patients are most likely to respond. A recent study indicates that a probiotic can modulate

D
HPA activity and elevate oxytocin levels. If this is replicated in humans it may be that social

behavior patterns can be altered via oxytocin by targeting gut microbes [43, 44].

TE
The gut microbiota in patients with autism spectrum disorder has been sequenced [45]. In the

most recently published study, Tomova et al studied the microbiota in Slovakian children

with autism. They found a significant decrease in the Bacteroidetes/Firmicutes ratio and
EP
elevation in Lactobacillus spp numbers. There was a modest elevation in Desulfovibrio spp

and a correlation with the severity of autism. A probiotic in the diet was found to normalise

the Bacteroidetes/Firmicutes ratio and Desulfovibrio spp levels. An analysis by Mayer &
C

colleagues concludes that there is a shortage of large comprehensive studies of the

microbiome in autism [12] and one can only agree with this assessment. Furthermore, the
C

issue of which comes first is fundamental; are these microbiota changes epiphenomenal,
A

induced by unusual diets seen in many individuals as a product of obsessional ruminations or

are they core to the disease pathophysiology? Many studies to date fail to adequately address

the heterogeneous aspect of this disorder. Future studies will need to do so if progress is to

be made. More comprehensive and accurate phenotyping is required.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Nutrition is clearly an important variable in any microbiota studies and especially in the

context of autism. This was clearly demonstrated in a microbiome composition study, which

showed an elevation in the low-abundance Cyanobacteria/chloroplast genus in children

diagnosed with ASD (Son et al., 2015). However, due to availability of dietary information it

was found that this was probably due to intake of chia seeds which are often present in

specialized diets.

D
Schizophrenia

TE
It has been argued that our social and cognitive development, in evolutionary terms, has been

aided by our symbiotic relationship with microbes. Disruption in social and cognitive

functioning is central to the pathophysiology of schizophrenia. Given this fact there has been
EP
recent focus on the microbiota in the disorder, though no large scale studies have yet been

published.
C

Toxoplasma gondii, the protozoa, is known to cause major changes to the gut microbiota

[46] and is an established environmental risk factor for schizophrenia [47]. In mice it can
C

radically alter behavior patterns resulting in reckless behavior in the presence of predators.
A

[48]. An intriguing human study reported that latent infection in healthy elderly can result

in deficits in goal directed learning, associated with underlying changes in dopaminergic

neural transmission [49].

In the elderly C. difficile infection can be treatment refractory and a few cases of

schizophrenia have been reported in association with C. difficile infection. Perhaps the

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
association is entirely coincidental, though it has been postulated as mediated by a

phenylalanine derivative synthetized by the bacteria [50]. C. difficile is also found in some

babies born by Caesarean section, in whom it does not create obvious difficulties. However,

there are no published studies available on the long-term outcome of such babies.

To date no systematic analysis of the gut microbiota in schizophrenia has taken place, though

it has been argued that any genomic analysis in the disorder should include an analysis of gut

D
microbial DNA [51].

TE
Psychobiotics

Fermented foods are a traditional part of the diet in many cultures and it is argued that such

intake decreases the levels of mood disorders[52]. For example, in Japan traditional dietary
EP
practice includes fermented soy products and this has been linked to lower rates of

depression[53].

Psychobiotics were first defined as ‘the family of bacteria that, ingested in appropriate

quantities, had a positive mental health benefit’ [54]. Recently, the definition has been
C

expanded to include prebiotics, which are dietary, soluble fibres, for example

galactooligosaccharides (GOS) or fructooligosaccharides (FOS), that stimulate the growth of


C

‘good’ bacteria.

There is an expanding volume of preclinical data to support the concept of psychobiotics.


A

Understandably, clinical data is less abundant but nonetheless is emerging. Given the

demonstrated efficacy of probiotics in IBS [55] and the high co-morbidity between IBS and

stress related mental health issues, such as anxiety and depression, it is not surprising that

certain probiotics might positively impact on mental health.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Bifidobacterium longum 1714 in a placebo-controlled study in healthy young volunteers not

only reduced stress responses but enhanced cognitive activity. Morning cortisol was

decreased on the psychobiotic relative to that seen with placebo. In marked contrast a

Lactobacillus rhamnosus (JB1 strain) failed in translation; preclinical studies found

significant neurochemical and behavioural effects[19], but no impact was found in a placebo-

controlled study in healthy volunteers[56].

Mayer’s group at UCLA [57] administered healthy female participants either a placebo or a

D
fermented dairy drink made from the probiotics (Bifidobacterium animalis Lactis,

Streptococcus thermophiles, Lactobacillus bulgaricus, and Lactococcus lactis Lactis), which

TE
were consumed over 4 weeks. All subjects underwent functional magnetic resonance imaging

(fMRI) to determine how probiotic ingestion affected brain function. Subjects were shown

emotional faces that are known to capture attention and cause brain activation. Relative to
EP
placebo, probiotic-treated participants showed decreased activity in a ‘functional network

associated with emotional, somatosensory, and interceptive processing, including the

somatosensory cortex, the insula, and the periaqueductal grey’. In marked contrast, placebo

treated subjects showed increased activity in these regions in response to same stimuli. The
C

authors interpret this as evidence of a probiotic-induced reduction in network-level neural

reactivity to negative emotional information. The study is noteworthy as the first to


C

demonstrate the capacity of probiotics to alter brain physiology.


A

A recent prebiotic study carried out in Oxford found a significant impact on stress responses

[58]. Healthy male and female participates consumed either BGOS, FOS, or a placebo. In

comparison to the other two groups, participants who consumed BGOS showed significantly

reduced waking-cortisol responses, which are a robust marker of anxiety, stress, and

depression risk [59]. Participants also completed an emotional dot-probe task measuring

vigilance, or attention to negative stimuli, which is a marker of anxiety and depression.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Participants taking BGOS showed substantially attenuated vigilance on this task, suggesting

reduced attention and reactivity to negative emotions. Overall, the data support the view that

the specific prebiotic has anxiolytic activity.

Another study of a psychobiotic on stress related parameters was carried out by Takada and

colleagues[60]. They conducted a placebo controlled trial of Lactobacillus casei strain

Shirota (LcS) exploring both psychological and physiological parameters. The study group

was healthy medical students who were participating in an exam. This is a well-established

D
stress paradigm. Subjects were randomised to receive either LcS-fermented milk or placebo

daily for 8 weeks followed by the exam. All subjects completed anxiety scores and provided

TE
saliva samples for cortisol measurement. Any side effects were carefully documented.

Academic stress resulted in increases in salivary cortisol and an increase in physical

symptoms, both of which were significantly suppressed in the LcS group. No significant side
EP
effects were reported.

In a parallel animal study, rats were fed a regular diet with or without LcS for 14 days. They

were then subjected to a water avoidance stress (WAS) test. Blood was drawn from a tail vein

for corticosterone measurement and the expression of cFos and corticotropin releasing factor
C

(CRF) in the paraventricular nucleus (PVN) were determined after the WAS test. In animals

treated with LcS, WAS-induced elevations in corticosterone were attenuated, and the number
C

of CRF-expressing neurons in the PVN was decreased. A mechanistic understanding of these


A

findings is provided by the fact that intra-gastric infusion of LcS stimulated gastric vagal

afferent activity and did so in a dose-dependent manner. The results suggest that LcS may

positively impact stress responses by acting through the vagus nerve.

A large scale cross-sectional study has examined the impact of probiotics on measures of

social anxiety [61]. Seven hundred and ten young adults completed measures of dietary

intake and personality traits. Controlling for relevant variables it was shown that exercise,

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
neuroticism, and fermented food consumption were a significantly predictor of social anxiety.

The data indicate that fermented food intake interacts with the personality trait of neuroticism

in predicting social anxiety. Those subjects with high neuroticism scores but a high frequency

of fermented food intake had lower symptoms of social anxiety. The data support the view

that fermented foods containing psychobiotics have a preventive effects against social anxiety

in those at high genetic risk.

Sternbergen et al [62] tested a polybiotic (multi-strain probiotic)containing Bifidobacterium

D
bifidum, Bifidobacterium lactis, Lactobacillus acidophilus, Lactobacillus brevis,

Lactobacillus casei, Lactobacillus salivarius, and Lactococcus lactis in normonthymic (non-

TE
depressed) individuals using a triple-blind, placebo-controlled, randomized, design, Twenty

healthy participants received 28 days of polybiotic treatment while 20 controls received an

inert placebo. Subjects who received the probiotics showed a statistically significant
EP
reduction in cognitive reactivity to sad mood. The results provide evidence that probiotics

may help reduce negative mood responses at least in healthy subjects.

Romijn and Rucklidge in their systematic review [63] add a note of caution to the above

optimistic findings concluding that more well conducted studies are required before any
C

definitive conclusions can be drawn about the efficacy of probiotics in mental illness.

Further studies of a translational nature are certainly required.


C
A

Defining dysbiosis

What is a ‘normal’ gut microbiota? We still cannot say with certainty what the ideal

composition of the gut microbiota is for optimal wellbeing. With the decreasing costs of

sequencing and the large scale studies currently underway this issue is likely to be resolved in

the not too distant future. At this point we have small scale studies in clinical populations,

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
that at best can be described as preliminary and from a bioinformatics perspective are

worryingly under-powered. We urgently require a profiling of the microbiota in the common

psychiatric disorders in large numbers of well phenotyped patients, who are preferably drug

naïve, in the early stages of their illness and whose nutritional status is well documented. We

are far from this point at present and therefore the extent to which a dysbiosis may play a role

in mental illness is far from certain.

D
Lost in translation?

TE
As the pharmaceutical industry has learned, therapeutic data acquired from rodent studies is

frequently lost in translation. There have been numerous false positive studies of compounds

which looked promising in animal models, but failed to achieve effects beyond those of
EP
placebo in clinical trials. In the development of psychobiotics, it is unclear whether a single

strain is preferable to the use of multiple strains or whether prebiotics which may influence a

wide range of bacteria may be more impacting than administering bacteria. Neither do we

know what the most effective in vitro screening techniques for detecting psychobiotics are.
C

One approach might be the use of in vitro assays to detect bacteria showing promising anti-
C

inflammatory activity. Bifidobacterium infantis 35624 which is effective in treating irritable

bowel syndrome was initially detected by such a screen. Another approach might be to
A

profile the metabolic output of bacteria by studying the content of the supernatants for SCFAs

and neurotransmitters. We know for example that most lactobacilli secrete the inhibitory

neurotransmitter GABA to varying extents. Are bacteria capable of improving gut barrier

function likely to have positive mental health benefits?

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
If psychobiotics for treating depression are to emerge the protection of intellectual property in

the field is essential. This is an evolving legal area and far more complex than the protection

of traditional small molecules. Spending large sums of money developing a psychobiotic,

only to find that a dietary supplement company market related bacteria with no efficacy data,

is unlikely to be attractive to big pharmaceutical companies. Appropriate regulation in this

area is of paramount importance to attract investment.

D
Perhaps the main attraction to industry is the potential for rapidly developing psychobiotics in

a manner not possible with small molecules. Bacteria which have GRAS status (generally

TE
regarded as safe) do not need time consuming and expensive toxicological analysis. This

conceivably means that the average 10 year development programme for an antidepressant

can be significantly shortened.


EP
As we documented in this review and elsewhere [64], research on microbiota has shown

promising results in preclinical models of psychiatric illness that may translate to patient care.

Will the next decade be for psychobiotics and the brain-gut-microbiota axis what the nineteen
C

fifties was in terms of antidepressant and antipsychotic development? Only time will tell if

the new paradigm is a fruitful one or a false dawn. However, what is certain is that this
C

paradigm is currently at an embryonic level of development, requiring major financial

investment if it is to progress.
A

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
References

1. Chertok L: [30 years later. The story of the discovery of neuroleptics]. Annales

medico-psychologiques 1982, 140(9):971-976.

2. Van De Wardt-Kikkert NM, Rentmeester J: [The use of largactil in psychiatry].

Geneeskundige gids 1953, 31(24):499-507.

3. Domino EF: History of modern psychopharmacology: a personal view with an

D
emphasis on antidepressants. Psychosomatic medicine 1999, 61(5):591-598.

4. Lopez-Munoz F, Alamo C: Monoaminergic neurotransmission: the history of the

TE
discovery of antidepressants from 1950s until today. Current pharmaceutical design

2009, 15(14):1563-1586.

5. Hirschfeld RM: History and evolution of the monoamine hypothesis of depression.


EP
The Journal of clinical psychiatry 2000, 61 Suppl 6:4-6.

6. Stanniland C, Taylor D: Tolerability of atypical antipsychotics. Drug safety 2000,

22(3):195-214.

7. Khoury B, Knauper B, Schlosser M, Carriere K, Chiesa A: Effectiveness of traditional


C

meditation retreats: A systematic review and meta-analysis. Journal of psychosomatic


C

research 2017, 92:16-25.

8. Coronado-Montoya S, Levis AW, Kwakkenbos L, Steele RJ, Turner EH, Thombs


A

BD: Reporting of Positive Results in Randomized Controlled Trials of Mindfulness-

Based Mental Health Interventions. PloS one 2016, 11(4):e0153220.

9. Lilja JL, Zelleroth C, Axberg U, Norlander T: Mindfulness-based cognitive therapy is

effective as relapse prevention for patients with recurrent depression in Scandinavian

primary health care. Scandinavian journal of psychology 2016.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
10. Cladder-Micus MB, Vrijsen JN, Becker ES, Donders R, Spijker J, Speckens AE: A

randomized controlled trial of Mindfulness-Based Cognitive Therapy (MBCT) versus

treatment-as-usual (TAU) for chronic, treatment-resistant depression: study protocol.

BMC psychiatry 2015, 15:275.

11. Furukawa TA, Noma H, Caldwell DM, Honyashiki M, Shinohara K, Imai H, Chen P,

Hunot V, Churchill R: Waiting list may be a nocebo condition in psychotherapy trials:

a contribution from network meta-analysis. Acta psychiatrica Scandinavica 2014,

D
130(3):181-192.

12. Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K: Gut microbes and the

13.
TE
brain: paradigm shift in neuroscience. The Journal of neuroscience : the official

journal of the Society for Neuroscience 2014, 34(46):15490-15496.

Dinan TG, Cryan JF: The impact of gut microbiota on brain and behaviour:
EP
implications for psychiatry. Current opinion in clinical nutrition and metabolic care

2015, 18(6):552-558.

14. Dinan TG, Cryan JF: Gut instincts: microbiota as a key regulator of brain

development, ageing and neurodegeneration. J Physiol 2017, 595(2):489-503.


C

15. Rhee SH, Pothoulakis C, Mayer EA: Principles and clinical implications of the brain-

gut-enteric microbiota axis. Nature reviews Gastroenterology & hepatology 2009,


C

6(5):306-314.
A

16. El Aidy S, Dinan TG, Cryan JF: Gut Microbiota: The Conductor in the Orchestra of

Immune-Neuroendocrine Communication. Clinical therapeutics 2015, 37(5):954-967.

17. Grenham S, Clarke G, Cryan JF, Dinan TG: Brain-gut-microbe communication in

health and disease. Frontiers in physiology 2011, 2:94.

18. Dinan TG, Stilling RM, Stanton C, Cryan JF: Collective unconscious: how gut

microbes shape human behavior. Journal of psychiatric research 2015, 63:1-9.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
19. Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG,

Bienenstock J, Cryan JF: Ingestion of Lactobacillus strain regulates emotional

behavior and central GABA receptor expression in a mouse via the vagus nerve.

Proceedings of the National Academy of Sciences of the United States of America

2011, 108(38):16050-16055.

20. Svensson E, Horvath-Puho E, Thomsen RW, Djurhuus JC, Pedersen L, Borghammer

P, Sorensen HT: Vagotomy and subsequent risk of Parkinson's disease. Annals of

D
neurology 2015, 78(4):522-529.

21. Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG: Effects of the

22.
TE
probiotic Bifidobacterium infantis in the maternal separation model of depression.

Neuroscience 2010, 170(4):1179-1188.

Lyte M: Microbial endocrinology in the microbiome-gut-brain axis: how bacterial


EP
production and utilization of neurochemicals influence behavior. PLoS pathogens

2013, 9(11):e1003726.

23. Lyte M: Microbial endocrinology and the microbiota-gut-brain axis. Advances in

experimental medicine and biology 2014, 817:3-24.


C

24. Stilling RM, Dinan TG, Cryan JF: Microbial genes, brain & behaviour - epigenetic

regulation of the gut-brain axis. Genes, brain, and behavior 2014, 13(1):69-86.
C

25. El Aidy S, Dinan TG, Cryan JF: Immune modulation of the brain-gut-microbe axis.
A

Frontiers in microbiology 2014, 5:146.

26. Capuron L, Hauser P, Hinze-Selch D, Miller AH, Neveu PJ: Treatment of cytokine-

induced depression. Brain, behavior, and immunity 2002, 16(5):575-580.

27. Abautret-Daly A, Dempsey E, Parra-Blanco A, Medina C, Harkin A: Gut-brain

actions underlying comorbid anxiety and depression associated with inflammatory

bowel disease. Acta neuropsychiatrica 2017:1-22.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
28. O'Mahony SM, Marchesi JR, Scully P, Codling C, Ceolho AM, Quigley EM, Cryan

JF, Dinan TG: Early life stress alters behavior, immunity, and microbiota in rats:

implications for irritable bowel syndrome and psychiatric illnesses. Biological

psychiatry 2009, 65(3):263-267.

29. Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, Wang W, Tang W, Tan Z, Shi J et

al: Altered fecal microbiota composition in patients with major depressive disorder.

Brain, behavior, and immunity 2015, 48:186-194.

D
30. Kelly JR, Borre Y, C OB, Patterson E, El Aidy S, Deane J, Kennedy PJ, Beers S,

Scott K, Moloney G et al: Transferring the blues: Depression-associated gut

31.
TE
microbiota induces neurobehavioural changes in the rat. Journal of psychiatric

research 2016, 82:109-118.

Lin H, Williams KA, Katsovich L, Findley DB, Grantz H, Lombroso PJ, King RA,
EP
Bessen DE, Johnson D, Kaplan EL et al: Streptococcal upper respiratory tract

infections and psychosocial stress predict future tic and obsessive-compulsive

symptom severity in children and adolescents with Tourette syndrome and obsessive-

compulsive disorder. Biological psychiatry 2010, 67(7):684-691.


C

32. Kantak PA, Bobrow DN, Nyby JG: Obsessive-compulsive-like behaviors in house

mice are attenuated by a probiotic (Lactobacillus rhamnosus GG). Behavioural


C

pharmacology 2014, 25(1):71-79.


A

33. Lyte M, Varcoe JJ, Bailey MT: Anxiogenic effect of subclinical bacterial infection in

mice in the absence of overt immune activation. Physiology & behavior 1998,

65(1):63-68.

34. Bruch JD: Intestinal infection associated with future onset of an anxiety disorder:

Results of a nationally representative study. Brain, behavior, and immunity 2016.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
35. Zahorodny W, Shenouda J, Howell S, Rosato NS, Peng B, Mehta U: Increasing

autism prevalence in metropolitan New Jersey. Autism : the international journal of

research and practice 2014, 18(2):117-126.

36. Li Q, Zhou JM: The microbiota-gut-brain axis and its potential therapeutic role in

autism spectrum disorder. Neuroscience 2016, 324:131-139.

37. Borre YE, O'Keeffe GW, Clarke G, Stanton C, Dinan TG, Cryan JF: Microbiota and

neurodevelopmental windows: implications for brain disorders. Trends in molecular

D
medicine 2014, 20(9):509-518.

38. Desbonnet L, Clarke G, Shanahan F, Dinan TG, Cryan JF: Microbiota is essential for

39.
TE
social development in the mouse. Molecular psychiatry 2014, 19(2):146-148.

Borre YE, Moloney RD, Clarke G, Dinan TG, Cryan JF: The impact of microbiota on

brain and behavior: mechanisms & therapeutic potential. Advances in experimental


EP
medicine and biology 2014, 817:373-403.

40. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Codelli JA, Chow

J, Reisman SE, Petrosino JF et al: Microbiota modulate behavioral and physiological

abnormalities associated with neurodevelopmental disorders. Cell 2013, 155(7):1451-


C

1463.

41. Chini B, Leonzino M, Braida D, Sala M: Learning about oxytocin: pharmacologic and
C

behavioral issues. Biological psychiatry 2014, 76(5):360-366.


A

42. Shen H: Neuroscience: The hard science of oxytocin. Nature 2015, 522(7557):410-

412.

43. Erdman SE, Poutahidis T: Probiotic 'glow of health': it's more than skin deep.

Beneficial microbes 2014, 5(2):109-119.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
44. Buffington SA, Di Prisco GV, Auchtung TA, Ajami NJ, Petrosino JF, Costa-Mattioli

M: Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic

Deficits in Offspring. Cell 2016, 165(7):1762-1775.

45. Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, Ostatnikova

D: Gastrointestinal microbiota in children with autism in Slovakia. Physiology &

behavior 2015, 138:179-187.

46. Molloy MJ, Grainger JR, Bouladoux N, Hand TW, Koo LY, Naik S, Quinones M,

D
Dzutsev AK, Gao JL, Trinchieri G et al: Intraluminal containment of commensal

outgrowth in the gut during infection-induced dysbiosis. Cell host & microbe 2013,

47.
14(3):318-328.

TE
Bhadra R, Cobb DA, Weiss LM, Khan IA: Psychiatric disorders in toxoplasma

seropositive patients--the CD8 connection. Schizophrenia bulletin 2013, 39(3):485-


EP
489.

48. Vyas A, Kim SK, Giacomini N, Boothroyd JC, Sapolsky RM: Behavioral changes

induced by Toxoplasma infection of rodents are highly specific to aversion of cat

odors. Proceedings of the National Academy of Sciences of the United States of


C

America 2007, 104(15):6442-6447.

49. Beste C, Getzmann S, Gajewski PD, Golka K, Falkenstein M: Latent Toxoplasma


C

gondii infection leads to deficits in goal-directed behavior in healthy elderly.


A

Neurobiology of aging 2014, 35(5):1037-1044.

50. Shaw W: Increased urinary excretion of a 3-(3-hydroxyphenyl)-3-hydroxypropionic

acid (HPHPA), an abnormal phenylalanine metabolite of Clostridia spp. in the

gastrointestinal tract, in urine samples from patients with autism and schizophrenia.

Nutritional neuroscience 2010, 13(3):135-143.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
51. Dinan TG, Borre YE, Cryan JF: Genomics of schizophrenia: time to consider the gut

microbiome? Molecular psychiatry 2014, 19(12):1252-1257.

52. Selhub EM, Logan AC, Bested AC: Fermented foods, microbiota, and mental health:

ancient practice meets nutritional psychiatry. Journal of physiological anthropology

2014, 33:2.

53. Nanri A, Kimura Y, Matsushita Y, Ohta M, Sato M, Mishima N, Sasaki S, Mizoue T:

Dietary patterns and depressive symptoms among Japanese men and women.

D
European journal of clinical nutrition 2010, 64(8):832-839.

54. Dinan TG, Stanton C, Cryan JF: Psychobiotics: a novel class of psychotropic.

55.
TE
Biological psychiatry 2013, 74(10):720-726.

Didari T, Mozaffari S, Nikfar S, Abdollahi M: Effectiveness of probiotics in irritable

bowel syndrome: Updated systematic review with meta-analysis. World journal of


EP
gastroenterology : WJG 2015, 21(10):3072-3084.

56. Kelly JR, Allen AP, Temko A, Hutch W, Kennedy PJ, Farid N, Murphy E, Boylan G,

Bienenstock J, Cryan JF et al: Lost in translation? The potential psychobiotic

Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in


C

healthy male subjects. Brain, behavior, and immunity 2017, 61:50-59.

57. Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, Guyonnet D, Legrain-


C

Raspaud S, Trotin B, Naliboff B et al: Consumption of fermented milk product with


A

probiotic modulates brain activity. Gastroenterology 2013, 144(7):1394-1401, 1401

e1391-1394.

58. Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PW: Prebiotic

intake reduces the waking cortisol response and alters emotional bias in healthy

volunteers. Psychopharmacology 2015, 232(10):1793-1801.

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
59. Bhagwagar Z, Hafizi S, Cowen PJ: Increased salivary cortisol after waking in

depression. Psychopharmacology 2005, 182(1):54-57.

60. Takada M, Nishida K, Kataoka-Kato A, Gondo Y, Ishikawa H, Suda K, Kawai M,

Hoshi R, Watanabe O, Igarashi T et al: Probiotic Lactobacillus casei strain Shirota

relieves stress-associated symptoms by modulating the gut-brain interaction in human

and animal models. Neurogastroenterology and motility : the official journal of the

European Gastrointestinal Motility Society 2016.

D
61. Hilimire MR, DeVylder JE, Forestell CA: Fermented foods, neuroticism, and social

anxiety: An interaction model. Psychiatry research 2015, 228(2):203-208.

62.

TE
Steenbergen L, Sellaro R, van Hemert S, Bosch JA, Colzato LS: A randomized

controlled trial to test the effect of multispecies probiotics on cognitive reactivity to

sad mood. Brain, behavior, and immunity 2015, 48:258-264.


EP
63. Romijn AR, Rucklidge JJ: Systematic review of evidence to support the theory of

psychobiotics. Nutrition reviews 2015, 73(10):675-693.

64. Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease.

Gastroenterol Clin North Am 2017;46:77–


C

89. http://dx.doi.org/10.1016/j.gtc.2016.09.007
C
A

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Figure Legend

The brain-gut-microbiota axis in health and disease. The routes of communicate between gut

and brain include neural, humoral and immune pathways. Gut dysbiois leads to altered

immunology, activation of the HPA, altered levels of short chain fatty acids and tryptophan,

together with aberrant signalling through the vagus nerve. Psychobiotics have the potential to

normalize such processes.

D
TE
EP
C
C
A

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.
Figure 1

D
TE
EP
C
C
A

Copyright © 2017 by the American Psychosomatic Society. Unauthorized reproduction of this article is prohibited.

You might also like