You are on page 1of 13

Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

Contents lists available at ScienceDirect

Neuroscience and Biobehavioral Reviews


journal homepage: www.elsevier.com/locate/neubiorev

Review

What’s bugging your teen?—The microbiota and adolescent mental


health
Karen-Anne McVey Neufeld a , Pauline Luczynski a , Clara Seira Oriach a,b ,
Timothy G. Dinan a,b , John F. Cryan a,c,∗
a
APC Microbiome Institute, University College Cork, Ireland
b
Department of Psychiatry, University College Cork, Ireland
c
Department of Anatomy and Neuroscience, University College Cork, Ireland

a r t i c l e i n f o a b s t r a c t

Article history: Human adolescence is a time of enormous developmental change, second only to infancy and early child-
Received 4 March 2016 hood in terms of brain shaping and growth. It is also a period in life when the young adult is faced with
Received in revised form 4 June 2016 distinct environmental challenges and stressors. Interestingly, we now know that these external sources
Accepted 6 June 2016
of stress all have an impact on the intestinal microbiota. Given that there is now a significant body of
Available online 7 June 2016
knowledge indicating a role for the microbiota-gut-brain axis in development and function of the brain,
and potentially the emergence of psychiatric illnesses, we need to draw our attention to the intestinal
Keywords:
microbiota in the adolescent. As psychiatric illnesses frequently first manifest during the teenage years it
Adolescence
Microbiota-gut-brain axis
may be that the intestinal bacteria are playing an as yet unidentified role in disease pathogenesis. Iden-
Development tifying a role for the microbiota in psychiatric illnesses opens up an exciting opportunity for therapeutic
Psychiatric illnesses advances via bacterial manipulation. This could prove to be a beneficial and novel avenue for treatment
Early life challenges of mental illnesses in the developing teen.
Probiotics © 2016 Elsevier Ltd. All rights reserved.
Hypothalamic-pituitary-adrenal axis
Brain plasticity
Critical windows

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
2. Microbiota-gut-brain axis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 301
3. Brain development – focus on adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
3.1. Structural brain development in adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302
3.2. Brain function in adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303
3.3. Stress responsivity in adolescence. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .303
4. Enteric nervous system – focus on development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
5. Microbiota development – focus on adolescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304
6. Adolescent mental health – what role could bacteria be playing? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 305
6.1. Stress-resilience and microbiota . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
6.2. Diet, alcohol and exercise – effects on the microbiota . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 306
7. Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
8. Concluding remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307

Abbreviations: ACTH, adrenocorticotropic hormone; AH, afterhyperpolarization; BDNF, brain-derived neurotrophic factor; CNS, central nervous system; ENS, enteric
nervous system; GF, germ-free; HPA, hypothalamic-pituitary-adrenal; IBS, irritable bowel syndrome; MIA, maternal immune activation; PUFA, polyunsaturated fatty acid;
VPA, valproic acid.
∗ Corresponding author at: APC Microbiome Institute, University College Cork, Ireland.
E-mail address: j.cryan@ucc.ie (J.F. Cryan).

http://dx.doi.org/10.1016/j.neubiorev.2016.06.005
0149-7634/© 2016 Elsevier Ltd. All rights reserved.
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 301

1. Introduction and well-being than previously considered, and that the benefits of
a synergistic relationship with bacteria can extend to human men-
As a society, we are well aware of the many and varied external tal health. Most interestingly, we now know that the microbiota
sources of pressure adolescents face; from possible drug and alco- influence the expression of host behavior and likely play a role in
hol exposure, to social issues involving peers, to changing habits pathophysiology of psychiatric disease (Foster and McVey Neufeld,
related to eating and sleeping, to balancing workload and the seem- 2013).
ingly inevitable associated stress. What we are likely less aware of Despite the fact that the concept of a microbiota-gut-brain axis
is that teenagers face additional developmental influence from the is now well established both pre-clinically and clinically, exact
inside – in the form of the commensal intestinal microbiota. In fact, mechanisms by which communication occurs are still under inves-
we now know that collectively extrinsic factors and the internal tigation. A number of systems are involved in this highway of
microbial environment work in concert to exert defining effects on information transfer, likely working in parallel to transmit infor-
host anatomy, physiology and behavior (Cryan and Dinan, 2012; mation between the microbiota and the brain, with neural (both
McVey Neufeld et al., 2013; Borre et al., 2014; De Palma et al., 2014; autonomic and enteric), immune, and endocrine pathways all
McVey Neufeld et al., 2016). In addition, emergence of psychiatric engaged in the constant crosstalk (Cryan and Dinan, 2012; Foster
illness is often first seen during the adolescent period when envi- and McVey Neufeld, 2013; El Aidy et al., 2015; Mayer et al., 2015).
ronmental stressors frequently peak (Paus et al., 2008; O’Connor The enteric nervous system (ENS) is a dedicated nervous system
and Cryan, 2014). Intriguingly, for all of the external influencing housed with the gastrointestinal wall that exists from the esopha-
factors listed above, lasting effects on the gut bacteria have been gus to the anus. While the ENS makes connections to the extrinsic
identified, leading to the hypothesis that mental illness emerging nervous system, it is also capable of operating independently of
during adolescence could in part be mediated by the commensal the spinal cord and brain (Costa et al., 2000). Both the ENS and the
intestinal microbiota (see Fig. 1). vagal nerve have proven to be important in transmitting informa-
The human gut houses 100 trillion bacteria, and the impact of tion regarding the intestinal microbiota from the gut to the brain,
our synergistic coexistence with these bugs on human develop- which is unsurprising when we consider that the first neural point
ment and function is only beginning to be appreciated (Backhed of contact for the gut bacteria is the approximately 500 million
et al., 2005; Frank and Pace, 2008). A recent study examining the neurons housed within the ENS and extending the full length of
variation in microbiota in healthy adults has identified a human the gastrointestinal tract (Furness, 2006; Blackshaw et al., 2007).
core microbiota that is globally observed, and is made up of 17 Pre-clinical studies carried out in mice have shown that the vagal
bacterial genera (Falony et al., 2016). In this review we will intro- nerve can be necessary for microbiota-gut-brain communication,
duce the concept of the microbiota-gut-brain axis, a bi-directional but these findings seem to be dependent on the bacterial species
axis of communication with broad implications for human physiol- in question. Both vagal dependence (Lyte et al., 2006; Bercik et al.,
ogy, health and disease. We will focus on the developing adolescent 2011b; Bravo et al., 2011) and independence (Bercik et al., 2010,
brain, its fragile balance between plasticity and vulnerability, and 2011a) have been demonstrated in rodent studies incorporating
the role that this axis, and the intestinal microbiota particularly, bacterial treatments with vagotomy.
may play in both its normal, healthy development and the patho- The immune system is unquestionably involved in microbiota-
physiology of the psychiatric illnesses that frequently first manifest gut-brain communication, but again the degree to which it is
during this critical window. We will discuss studies examining the necessary for the transmission of specific information seems to
use of both pre- and pro-biotics; the latter are live bacteria that con- depend upon the bacterial species under investigation. Early work
fer health benefits to the host and which only transiently inhabit the has demonstrated that sub-clinical doses of pathogenic bacteria
gut, and prebiotics are the indigestible foods (primarily carbohy- administered to mice could increase anxiety-like behavior in the
drates), that selectively promote the growth of certain gut bacteria, absence of changes to peripheral cytokine levels (Lyte et al., 2006). A
thereby providing indirect health benefits to the host (Gibson and more recent study in immunocompromised animals demonstrated
Roberfroid, 1995; Roberfroid, 2007). Finally, we will propose poten- that B and T cell deficient Rag1 knockout mice, which have altered
tial lines of inquiry for future research on therapeutics aimed at neurological and gut function, show normalization of some deficits
the treatment of psychiatric illnesses via the intestinal commensal following probiotic treatment (Smith et al., 2014).
microbiota. Research using germ-free (GF) mice, animals raised and main-
tained in the total absence of bacteria, has provided perhaps the
most persuasive evidence for a role of the microbiota in brain-gut
2. Microbiota-gut-brain axis signalling. GF animals show significantly altered immune, gastroin-
testinal, digestive, and metabolic function (for review see Luczynski
The existence of a gut-brain axis has been acknowledged for et al., 2016). Moreover, the absence of microbes during develop-
decades, with both clinical and basic research identifying this bidi- ment dramatically affects the brain-gut axis and central nervous
rectional axis of communication as fundamental for both normal system (CNS) circuitry and wiring, although exact mechanisms
gastrointestinal function but also to the frequently co-morbid psy- whereby these changes occur remain unknown. Indeed, GF ani-
chiatric and bowel diseases (Mayer, 2000). In the last decade, the mals show anxiolytic-like behavior (Diaz Heijtz et al., 2011; Neufeld
concept of this axis has been extended to the “microbiota-gut-brain et al., 2011; Clarke et al., 2013), reduced sociability (Desbonnet
axis”, a reflection of our increased understanding of the impor- et al., 2014; Arentsen et al., 2015) and learning deficits (Gareau
tance of the trillions of bugs residing and working in the human et al., 2011). GF mice also demonstrate heightened hypothalamic-
gut (Rhee et al., 2009; Bercik, 2011; Cryan and O’Mahony, 2011). pituitary-adrenal (HPA) axis reactivity following exposure to a
Our relationship with the commensal bacteria is mutualistic – the stressor (Sudo et al., 2004), with stress hyperresponsivity known
bugs benefit from a rich and protected habitat, while we humans to induce CNS change. Most recently, it has been shown that GF
benefit as bacteria breakdown otherwise indigestible food prod- animals have differences in brain structure, with hypermyelina-
ucts, providing us with previously inaccessible nutrients. We also tion observed in the prefrontal cortex (Hoban et al., 2016), and
gain as these beneficial bacteria provide a biofilm through which it also marked differences in microglial cells (Erny et al., 2015). GF
is difficult for pathogenic bacteria to gain access. However, we are mice have more microglia throughout the brain compared to con-
beginning to understand that this mutually beneficial relationship trol mice, and these cells are clearly abnormal, with longer, more
has further far-reaching consequences for optimal human health complex processes. In addition, the microglia of GF mice do not dis-
302 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

Fig. 1. The microbiota-gut-brain axis in adolescence. This bidirectional axis is generally healthy and functioning, but environmental factors frequently encountered by the
developing teen – such as drugs of abuse, alcohol, altered nutrition and sleep, and stress all have known effects on the intestinal microbiota. Here we propose that these
environmentally induced alterations to the microbiota may be playing a role in psychiatric disease pathogenesis, particularly in relation to those disorders that often first
manifest in the teenage years.

play activated morphology following a bacterial or viral challenge, to leave the safety of home and thus gain independence from care-
indicating that the lack of microbiota somehow impact the host’s givers (Burnett et al., 2011; Spear, 2011). Refinement in social and
ability to mount appropriate immune responses both peripherally, cognitive abilities during adolescence also facilitates this transi-
and in the CNS (Erny et al., 2015). Interestingly, antibiotic-treated tion into adulthood (Burnett et al., 2011; Spear, 2011; Crone and
mice display an immature microglial profile similar to that of Dahl, 2012; Spear, 2013). Until recently, it was thought that the
GF mice (Erny et al., 2015). It is important to mention that the brain was finished developing at puberty. However, it is now clear
GF model is often criticized for its lack of clinical relevance, as that adolescence is a robust transitory period characterized by neu-
humans are never in a GF state (Funkhouser and Bordenstein, 2013). roanatomical change and maturation. These structural changes to
However, the use of GF animals in research is not to directly imi- the brain map onto behaviours commonly associated with ado-
tate the human condition. Instead, the GF model allows the study lescence, such as stress responsivity, cognitive function, reward
of the dysfunction which arises in the absence of any microbial sensitivity, and social cognition (Burnett et al., 2011; Spear, 2013).
input on host physiology and behavior. As GF animals are not
exposed to microbes from conception on, they are perhaps not
the ideal model to study experimental questions regarding the 3.1. Structural brain development in adolescence
effect of altered microbial composition occurring later in life. For
these reasons, alternative models such as antibiotic treatment must In the late 1960’s and 70’s research on post-mortem human
be explored further. Interestingly, many of the characteristics of brains revealed that certain regions, namely the frontal cortex, con-
GF mice (ie reduced anxiety-like behaviour, cognitive dysfunc- tinue to develop morphologically beyond early life and even the
tion, social deficits, microglia activation deficits, BDNF expression pubertal period (Huttenlocher, 1979; Huttenlocher et al., 1982;
changes) also emerge after a sustained antibiotic-induced deple- Benes, 1989). The two most salient structural alterations in the
tion of the microbiota (Desbonnet et al., 2015; Erny et al., 2015; adolescent frontal cortex are changes in synaptic connectivity
Bercik, 2011). and axonal myelination. The frontal cortex is involved in exec-
From infancy to old age, diet is one of the most important factors utive function, cognition, and memory (Miller and Cohen, 2001;
mediating the establishment and composition of the gut microbiota Tamminga and Buchsbaum, 2004). As such, it is unsurprising that
(Power et al., 2014). Correspondingly, the microbiota is essential this neuroanatomical development during adolescence occurs in
for the optimal digestion and vitamin absorption of the food we parallel with dramatic changes in identity, self-consciousness, and
eat (Flint et al., 2012). As previously mentioned, there is strong evi- cognitive flexibility (Burnett et al., 2011).
dence linking the composition of the gut microbiota to behavioral Early in postnatal life, the brain generates many new synaptic
alterations. Given that the diet can affect the gut microbiota, host connections between neurons in a process known as synapto-
nutrition is one of the most potent factors contributing to brain-gut genesis (Rakic et al., 1994; Huttenlocher and Dabholkar, 1997).
signaling and behavior (for review see Seira Oriach et al., 2016). This proliferation of synapses is then followed by a period of
Clearly neural, immune and endocrine systems are all respon- synaptic elimination or pruning. Synaptic pruning is an experience-
sive to changes in the intestinal microbiota and likely have dependent process in which the neural wiring in the brain is
overlapping roles in communicating information from the gut to fine-tuned: frequently used connections are strengthened and
the brain and vice versa, but we are still in the relative infancy of scarcely used connections are eradicated (Holtmaat and Svoboda,
understanding the exact mechanisms of activity for the microbiota- 2009). Although the exact mechanisms underlying this process
gut-brain axis. Furthermore, there is little to no research on the remain to be elucidated, it is understood that resident microglia
development of this axis over the lifespan, much less in the adoles- in the brain regulate the elimination of certain synapses and the
cent. What is abundantly clear is that there is cross-communication strengthening and maintenance of others (Hua and Smith, 2004;
between the systems; the subsequent sections of this review will Schafer et al., 2012). Moreover, recent evidence has implicated
detail what we know about adolescent development and changes immune signaling, specifically activation of the complement sys-
occurring centrally in the brain, as well as peripherally, in the tem, in the elimination of synapses during postnatal development
enteric nervous system and the gut microbiota. (Sekar et al., 2016). In sensory regions, synaptic density is highest
in the first months of life after which time neural connections begin
to be pruned, reaching adult levels before or around adolescence
3. Brain development – focus on adolescence (Cragg, 1975; Huttenlocher et al., 1982; Bourgeois and Rakic, 1993).
Structural development in the prefrontal cortex however, follows a
From an evolutionary point of view, the physiological changes strikingly different time course. Here, there is a proliferation in neu-
associated with adolescence are thought to prepare the young adult ral connectivity during childhood and again in puberty, followed
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 303

by a post-pubertal period of stability and a subsequent elimination adolescents typically behave more impulsively, as measured by
and reorganization of synaptic connections (Huttenlocher, 1979; delayed discounting (Olson et al., 2007). These changes in reward
Bourgeois et al., 1994; Woo et al., 1997). Therefore, during the tran- responsivity could be due to altered dopaminergic signalling in
sition from adolescence to adulthood, prefrontal cortical synapses reward-relevant subcortical regions. Indeed, there are profound
are systematically pruned resulting in a net decrease in synaptic changes in both dopamine levels and the expression of its receptor
density in this brain area. in areas implicated in reward seeking during adolescence in the rat
The second defining change in brain structure in the post- (Tarazi and Baldessarini, 2000).
pubertal period relates to myelination. During neural development, Social interactions with peers take on a significant importance
oligodendrocytes produce a layer of myelin, which wraps around in adolescence, with adolescents spending up to one third of their
axons. This process forms an insulating sheath, which increases waking time interacting with peers during the academic year
the speed at which neurons can transmit information. By the (Csikszentmihalyi et al., 1977; Spear, 2000). These experiences are
first few years of life, axons in the sensory and motor cortices thought to help develop the social skills necessary to achieve inde-
are already fully myelinated (Yakovlev and Lecours, 1967). In pendence from the home environment (Spear, 2000). However, this
contrast, myelination continues in the frontal cortex well into ado- is also a time in which adolescents can conform to their peers and
lescence (Benes, 1989). This increase in myelination accelerates the develop antisocial behaviors such as stealing, aggression, trespass-
speed and efficiency of neural communication across distant brain ing and cheating (Berndt and Keefe, 1979; Cairns et al., 1989). While
regions, impacting the functional connectivity of the adolescent it is impossible to tease apart all of the factors influencing the devel-
brain (Markham and Greenough, 2004; Fair et al., 2008). opment of social cognition during adolescence, pubertal hormones
Non-invasive brain imaging has provided further evidence of and neurodevelopment both are likely to play major roles (Spear,
the ongoing morphological development of the frontal cortex of 2000; Blakemore, 2008; Burnett et al., 2011; Spear, 2013; Brizio
adolescents. Cortical gray matter volume progressively increases et al., 2015). Although there has been relatively little research into
in the frontal and parietal lobes throughout childhood, peaking in the neural circuitry underlying the development of social cognition
puberty, and then begins to decrease (Pfefferbaum et al., 1994; in adolescence, imaging studies have provided evidence that there
Giedd et al., 1996; Reiss et al., 1996; Giedd et al., 1999; Sowell exists a network of brain areas associated with social cognition,
et al., 2001; Sowell et al., 2003; Giorgio et al., 2010). In contrast, and in particular the ability to mentalize (Frith and Frith, 2003;
white matter volume in these brain areas consistently increases Burnett et al., 2011; Brizio et al., 2015). This network is thought
throughout childhood and adolescence (Giedd et al., 1999; Barnea- to be the neural substrate for recognizing another person’s mental
Goraly et al., 2005). These changes in gray and white matter are state, including emotions, desires and beliefs (Frith and Frith, 2003).
thought to be due to the aforementioned changes in synaptic prun- This ‘social brain’ includes the prefrontal cortex and the amygdala,
ing and myelination (Blakemore and Choudhury, 2006; Paus et al., amongst other structures (Blakemore, 2008), and the develop-
2008; Tau and Peterson, 2010). The concomitant loss of gray matter ment of social cognition in adolescence may be related to the
and increase in white matter during adolescence is potentially due continued neurodevelopment of these brain regions (Blakemore
to increased axonal myelination (Sowell et al., 2003; Paus et al., and Choudhury, 2006; Paus et al., 2008; Tau and Peterson, 2010;
2008). Another interpretation is that changes in gray matter vol- Goddings et al., 2014, see Kilford et al., 2016).
ume reflect alterations in synaptic connectivity: that the increase Although most of the research on adolescent brain function has
and decrease of gray matter before and after puberty are a result of focused on the cortex, limbic regions are also involved in medi-
synaptic proliferation and elimination, respectively (Huttenlocher ating the emotional changes which occur during this period. The
and Dabholkar, 1997; Paus et al., 2008; Tau and Peterson, 2010). amygdala, in particular, is a key mediator of emotion and fear
(LeDoux, 2007). To investigate the neural correlates of the height-
3.2. Brain function in adolescence ened emotionality and peer focus of adolescents, developmental
studies often assess amygdalar activation in response to emotional
Developmental immaturities in cognitive control, attentional social stimuli (Spear, 2013). Adolescents sometimes (Monk et al.,
regulation, and response inhibition have been widely attributed to 2003; Hare et al., 2008), but not always (Pine et al., 2001), display
the delayed maturation of the frontal cortex (Casey et al., 2008; an increase in amygdala activity in response to emotional face cues.
Spear, 2013). Although adolescents can generally perform well These findings can potentially explain the reduced capacity to make
on cognitive tasks, performance deficiencies emerge when task rational decisions in stressful or emotional situations (Dahl, 2004;
demands increase or when emotions are heightened (Liston et al., Spear, 2013). Interestingly, social peers are effective in provoking
2009). These results can perhaps be explained by the decrease these heightened emotional states during adolescence, with ado-
in frontal brain activity and the increase in the involvement of lescents engaging in more risk-taking behaviour than adults when
emotional centres in adolescents when emotions or stress are par- tested in the presence of peers (Gardner and Steinberg, 2005). These
ticularly high (Casey et al., 2008). The development of the frontal results are particularly important given that adolescents often par-
cortex in adolescence is therefore thought to underlie the delay ticipate in risky behaviours in social situations (Steinberg, 2008).
in maturation of the ‘top-down’ control systems controlling sub-
cortical ‘bottom-up’ systems, which mediate emotions and reward 3.3. Stress responsivity in adolescence
(Casey et al., 2008; Spear, 2013).
Novel stimuli, risky situations, and drugs of abuse feed into Changes in endocrine signalling are a hallmark of adolescent
the brain’s reward circuitry, which is critical for seeking natural development. Although the majority of studies have investi-
rewards essential for survival (Nesse and Berridge, 1997). Ado- gated gonadal hormones, recent attention has focused on the
lescents typically (Galvan et al., 2006; Cohen et al., 2010; Van development of HPA axis signalling and stress reactivity dur-
Leijenhorst et al., 2010), although not always (Bjork et al., 2004), ing adolescence (for reviews see Romeo and McEwen, 2006;
show altered striatal responses to rewards. Moreover, reward seek- McCormick and Mathews, 2007; Romeo, 2010; Eiland and Romeo,
ing peaks in mid-adolescence and then gradually decreases into 2013; see Romeo et al., 2016). Our current knowledge of these
adulthood (Steinberg et al., 2009; Cauffman et al., 2010). Although pubertal stress responses has largely been gleaned from rodent
multiple factors impact risk-taking during adolescence, impulsive models. During puberty, gonadal hormones increase considerably,
behavior is likely a key mediator (Pfefferbaum and Wood, 1994). while basal levels of stress hormones remain comparatively sta-
In fact, impulsivity is associated with increased risk-taking and ble (Pignatelli et al., 2006). In contrast, stress-induced release
304 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

of corticosterone is prolonged in pre-pubertal versus adult rats adulthood, it also provides a viable route for information to travel
(Goldman et al., 1973; Vazquez and Akil, 1993; Romeo et al., 2004). from the intestinal microbiota to the brain.
Importantly, this elongated stress hormone release is not influ-
enced by the different gonadal hormonal backgrounds of rats in
these two stages of development (Romeo et al., 2004). Adult rats 5. Microbiota development – focus on adolescence
habituate to repeated responses to the same stressor; however,
pre-pubertal males, but not females, show heightened HPA axis The advent of next-generation microbial sequencing technology
signalling in response to a repeated stressor (Romeo et al., 2006). has changed what we know about the constituent bacteria in the
In agreement with these results, pre-pubertal rats exposed to a human gut and how it changes over the course of life (Dominguez-
stressor display increased anxiety- and depressive-like behaviours Bello et al., 2011; Forde and O’Toole, 2013). It was largely believed
(Vidal et al., 2007; Leussis and Andersen, 2008; McCormick that the fetus existed in a sterile environment, but while we now
et al., 2008). Taken together, these data clearly indicate that HPA know that we were incorrect in thinking that the human is GF while
axis signalling and stress reactivity undergo significant change in utero, it is still understood that colonization of the gut primarily
during adolescence; however, there remains a paucity of informa- occurs during birth and the subsequent immediate postnatal period
tion describing the mechanisms underlying these developmental (Funkhouser and Bordenstein, 2013). A variety of factors are known
alterations. to influence this early pattern of bacterial colonization, including
mode of delivery, (i.e. vaginal vs Caesarean), location of birth (i.e.
hospital vs homebirth), formula vs breastfeeding, and whether the
4. Enteric nervous system – focus on development mother or infant undergoes any antibiotic treatment in early life
(Stark and Lee, 1982; Penders et al., 2006; Adlerberth and Wold,
While there is admittedly little information on ENS changes 2009; Dominguez-Bello et al., 2010; Fallani et al., 2010; Le Huerou-
specific to adolescence, we do know that it continues to be sig- Luron et al., 2010; Roger et al., 2010; Bezirtzoglou et al., 2011;
nificantly plastic well into adulthood (Galligan et al., 1989; Giaroni Hussey et al., 2011; Azad et al., 2013; Prince et al., 2014; Arboleya
et al., 1999). Moreover, evidence shows that the ENS is receptive to et al., 2015; Mueller et al., 2015).
changing intestinal microbial status, with altered neuronal physi- Following the immediate postnatal period, we also now have
ology observed in response to bacteria, as well as changes observed more reliable information regarding the bacterial composition of
in mRNA gene expression in adult enteric neurons after introduc- the infant gut. In comparison to the adult, the infant microbiota is
tion of bacteria. Work examining GF mice that have been colonized much more simple and unstable (Fouhy et al., 2012; Borre et al.,
with commensal intestinal microbiota in adulthood shows that 2014). Over the course of the child’s lifetime we see changes in
intracellular recordings taken from sensory neurons residing in the resident intestinal microbiota, with by and large less species
the gut wall, the so-called afterhyperpolarization (AH) neurons, diversity than that of an adult (Ringel-Kulka et al., 2013). How-
are different from those obtained from adult GF animals. Specif- ever, as the child enters into the adolescent period the outwardly
ically, AH neurons are less excitable in GF mice, whereas if the observable growth and change underway is mirrored inside the
mouse has been “conventionalized” in adulthood with bacteria, intestine. In a study examining bacterial diversity across the life-
neurons respond with normal levels of excitability when com- time, researchers examined 3 age cohorts; young children (prior
pared to controls (McVey Neufeld et al., 2013). These same neurons to 7 years), young adults and the elderly. In so doing, the investi-
respond with increased excitability when exposed to probiotic Lac- gators were able to track microbial changes with increasing age,
tobacillus rhamnosus (JB1) (Kunze et al., 2009). Interestingly, these corresponding to gradual decreases in the number of aerobic and
sensory neurons have been shown to form a nicotinic synapse with facultative anaerobic bacteria, and increases in anaerobic species
vagal afferents leading to the brain (Perez-Burgos et al., 2014), (Hopkins et al., 2002). A high-throughput analysis of distal gut
and similarly, mixed mesenteric afferent recordings from adult microbiota comparing that of adolescents and adults found that
conventionalized-GF mice show levels of activity comparable to while there was a significantly higher amount of Bifidobacterium
controls, whereas recordings from these nerves in GF mice again and Clostridium genera in the younger cohort, actual species num-
demonstrate reduced excitability (McVey Neufeld et al., 2015). ber was similar in the two age groups (Agans et al., 2011). A more
Mixed nerve recordings also show increased activity after expo- recent study has compared the microbiota of pre-adolescent chil-
sure to probiotic JB-1 (Perez-Burgos et al., 2013). Levels of mRNA dren to that of adults and found that again, the younger cohort
gene expression of calcium binding protein calbindin in the enteric had higher numbers of Bifidobacterium, while adults demonstrated
neurons are also similar when comparing conventionalized-GF gut more Bacteroides (Hollister et al., 2015). In addition, the authors
tissue to that of controls, whereas GF animals show significantly note that they also observed differences in microbiota profiles from
less expression compared to both (McVey Neufeld et al., 2015). a functional perspective, with the microbiota found in children
Again, this illustrates that ENS tissue in the adult mouse is plastic enriched in functions related to growth and development, while the
and responds to changes in intestinal luminal status, and given that adult’s microbial communities demonstrated higher functionality
activity of these sensory AH neurons is transmitted to the brain via associated with inflammation and obesity (Hollister et al., 2015). It
the vagal nerve this could then provide an avenue whereby infor- should be noted that more studies are needed in order to examine
mation regarding the bacterial contents of the gut can be conveyed the development and change of intestinal microbiota over the lifes-
to the brain. While these results are from adult mouse tissue, it pan; those that have been done to date focus on the extremes of age
is entirely reasonable to assume that adolescent tissue would be (i.e. composition of microbiota during infancy and in the elderly),
similarly plastic and responsive to bacterial input, indicating that and instead we need more information that is reflective of bacterial
information travelling via the microbiota-gut-brain axis would be diversity over the normal, gradual transitions of maturation.
altered in adolescence depending on environmental, and subse- One of the more obvious changes associated with puberty
quent microbial, changes. This confirmation of previous findings of relates to sexually dimorphic development. After a relatively quies-
extended plasticity in the ENS (Galligan et al., 1989; Giaroni et al., cent period in juvenility, the hypothalamic-pituitary-gonadal axis
1999), compared to what has been reported in terms of central reactivates in adolescence, and continues to be active throughout
nervous tissue plasticity in response to adult commensal conven- adulthood (Marceau et al., 2015). This increased release of gonadal
tionalization, is very exciting. Not only does it indicate a potential hormones during adolescence exerts organizational and activa-
target of action for microbial based therapies that could extend into tional effects on the physical, behavioral, and emotional aspects
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 305

of development (Romeo, 2003). In particular, the brain under- Vrieze et al., 2012; Karlsson et al., 2013), and cardiovascular disease
goes sexual differentiation during puberty and adolescence, with (Koren et al., 2011; Howitt and Garrett, 2012; Karlsson et al., 2012;
ovarian and testicular hormones mediating the feminization and Tuohy et al., 2014; Hansen et al., 2015). Clinical studies linking psy-
masculinization of the brain, respectively (for review see Juraska chiatric disorders with intestinal microbiota have been slower in
et al., 2013). Intriguingly, changes in gut microbiota in mice also coming, but a significant body of preclinical literature now exists.
seem to follow a sexually divergent trajectory (Jasarevic et al., Preclinical work has either focused on administering beneficial (i.e.
2016). Sex-related differences in the adult intestinal profiles of mice probiotic), or pathogenic bacteria to a host, or manipulating the nor-
are abolished when male mice undergo castration (Yurkovetskiy mal commensal intestinal microbiota of a host (as in antibiotic or GF
et al., 2013). However, the mechanisms by which testosterone may studies). Studies infecting mice with subclinical doses of pathogenic
be affecting the commensal microbial community is still unclear. bacterial strains like Campylobacter jejuni and Citrobacter rodentium
Conversely, it appears that early-life microbial exposure itself can have shown induction of anxiety-like behavior and learning deficits
influence gonadal hormone levels the risk of autoimmune dis- in the absence of overt inflammatory responses (Lyte et al., 1998;
ease (Markle et al., 2013). Female non-obese diabetic mice are Lyte et al., 2006; Goehler et al., 2008; Gareau et al., 2011). Alter-
more susceptible to type 1 diabetes than males. However, colo- nately, administering probiotics, which are live organisms known
nization of female GF non-obese diabetic mice with the microbiota to confer health benefits on the recipient when ingested in ade-
of males conferred protection against diabetes pathology through quate amounts, have been demonstrated to have a beneficial effect
a testosterone-dependent mechanism (Markle et al., 2013). Taken on brain function and behavior. Both Bifidobacteria and Lactobacil-
together, these findings suggest that the microbiota and gonadal lus genera have demonstrated anxiolytic effects in both humans
hormones bidirectionally modulate each other. Given the resur- and mice (Bravo et al., 2011; Messaoudi et al., 2011b), and the Lac-
gence of gonadal hormone signaling in adolescence, more research tobacillus strains particularly have been shown to normalize HPA
is clearly needed to determine how these hormones impact the axis reponsivity, reverse stress-induced colonic motility, as well as
microbiota and vice versa. confer cognitive benefits in mice (Savignac et al., 2015). Several
Admittedly, we are still far off from an exhaustive list of changes rodent studies have shown that bacterial strains can ameliorate
that occur over the developing lifespan in the colonizing intestinal stress hyperresponsivity, cognitive dysfunction, visceral hypersen-
microbiota, but it is still quite clear that the bacteria in the gut nat- sitivity and anxiety-like behaviour in studies of chemically-induced
urally diversify throughout life and respond to physiological and colitis, infection, and early life adverse events (Gareau et al., 2007;
environmental cues varying from individual to individual. As men- O’Mahony et al., 2009; Bercik et al., 2010; Bercik et al., 2011b).
tioned previously, we know that the intestinal microbiota alter in There are still remarkably few studies examining the
response to many of the environmental stressors known to par- microbiota-gut-brain axis over the course of development. The
ticularly affect teens. Diet and sleep variations, exposure to drugs information that we do have can be gleaned from those studies
of abuse and alcohol, stress and extended antibiotic usage all affect examining the impact of colonizing adolescent GF mice with nor-
the composition of the constituent microbiota. Given that teenagers mal commensal bacteria, as well as those looking at the result
can be exposed to any combination of these stressors over the of antibiotic treatment in otherwise healthy juvenile rodents. As
adolescent period we are left questioning what role changes in previously described, GF mice have an abnormal behavioural and
intestinal microbial status may have on the development of ado- physiological profile and distinct alterations in HPA axis activity
lescent psychiatric disturbances. (Luczynski et al., 2016). Specifically, GF mice show anxiolytic-like
behavior, decreased social preference and discrimination, and cog-
nitive deficits (Sudo et al., 2004; Diaz Heijtz et al., 2011; Neufeld
6. Adolescent mental health – what role could bacteria be et al., 2011; Clarke et al., 2013; Desbonnet et al., 2014). In addi-
playing? tion, they show lifelong stress hyperreactivity, and significantly
increased plasma adrenocorticotropin hormone (ACTH) and cor-
Many psychiatric disorders first become evident during the ticosterone after stress exposure (Sudo et al., 2004). Interestingly,
adolescent years. Symptoms of anxiety, depression, psychosis, it has been demonstrated that if GF mice are colonized with com-
substance abuse and eating disorders often manifest at this crit- mensal bacteria by the adolescent period (in mice roughly equal
ical window of development (Paus et al., 2008; Sturman and to 4 weeks of age), their stress hyperresponsivity can be reversed.
Moghaddam, 2011; Jaworska and MacQueen, 2015), with a large This is not true however, if the bacteria are introduced in adulthood.
scale National Comorbidity Study conducted in the United States This finding is highly provocative, because it indicates that there is a
from 2001 to 2003 demonstrating that the peak age of onset for critical window of development, up to and including adolescence,
psychiatric disease is 14 years (Kessler et al., 2005). We are now whereby the gut bacteria can program central stress circuitry. A
beginning to understand that this is because the pathophysiology critical component of all of the psychiatric illnesses that often first
underpinning these disorders is strongly linked to the types of brain appear in adolescence is dysregulation of stress circuitry. It is there-
development that are occurring during this time. Even in healthy fore compelling to note that these same circuits are responsive to
adolescents without clinical psychiatric illness, we see changes to changes in gut microbial status at this window in development.
behavior related to increased risk taking (Spear, 2000; Steinberg, Antibiotic studies have proven a useful source of information
2008), sensation seeking (Giedd, 2009) impulsivity (Steinberg, in unraveling the role of the microbiota-gut-brain axis in behavior.
2010) and emotional instability (Spear, 2000). Alterations to these Administering antibiotics to rodents in order to deplete the micro-
neurodevelopmental pathways can occur due to the particular biota has resulted in a reduction in anxiety-like and exploratory
types of environmental challenges that the adolescent typically behavior that reverses after a two-week washout period (Bercik
faces, and intriguingly the intestinal microbiota have likewise been et al., 2011a). In addition, feeding adult mice an antibiotic cock-
demonstrated to be affected by these challenges. tail has recently been shown to result in cognitive dysfunction
The links between the importance of a stable gut microbiota as observed in the novel object recognition test, with associated
and human health have been most clearly illustrated in disorders changes to mRNA gene expression of brain-derived neurotrophic
primarily affecting peripheral systems such as irritable bowel syn- factor (BDNF) in both the hippocampus and prefrontal cortex
drome (IBS) (Salonen et al., 2010; Tana et al., 2010; Jeffery et al., (Frohlich et al., 2016). A more recent study examining the effects
2012; Wu, 2012), obesity (Ley et al., 2006; Turnbaugh et al., 2009; of antibiotics particularly during the adolescent period in mice
Tremaroli and Backhed, 2012), type 2 diabetes (Qin et al., 2012; found significant changes to adult behavior, brain neurochemistry,
306 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

and intestinal microbial content when compared to that of con- ity (Golubeva et al., 2015). One potential mechanism explaining
trols (Desbonnet et al., 2015). At weaning (3 weeks of age) mice these results is that stress alters the mother’s vaginal microbiome,
were given an antibiotic cocktail in drinking water over a 4 week which then results in a decreased transmission of certain bacterial
period, and then behavior was assessed in adulthood. Antibiotic strains to the offspring (Jasarevic et al., 2015). Rodent models of
treated mice showed anxiolytic-like behavior, altered cognitive stress in early life and adulthood also demonstrate altered com-
function in the novel object recognition test in that antibiotic position of the microbiota, immune dysfunction, and changes to
treated mice were unable to successfully discriminate between sociability behaviours and stress reactivity (O’Mahony et al., 2009;
a novel object and a familiar, and social behavior dysfunction De Palma et al., 2015). Importantly, the degree to which animals
in the social transmission of food preference test. Treated mice are affected by stress can be predicted by changes in the microbial
also showed significantly increased serum tryptophan and reduced composition (Bailey et al., 2011; Golubeva et al., 2015; Bharwani
kynurenine levels. In the brain, antibiotic treatment resulted in et al., 2016). These findings suggest that the microbiota could be
significantly reduced hippocampal BDNF mRNA, and reduced vaso- driving maladaptive responses to stress. Indeed, the microbiota
pressin mRNA in the hypothalamus. Alterations were also observed is required for the stress-induced induction of certain anxiety-
in brain monoamines and monoamine metabolites (Desbonnet like and depressive-like behaviours (De Palma et al., 2015). This
et al., 2015). This important study is the first to show that antibiotic raises the intriguing possibility that the composition of the micro-
treatment during adolescence in mice can result in long term alter- biota could be a susceptibility factor for responses to various
ations to brain function and gene expression, and clearly indicates stressful insults, particularly at vulnerable neurodevelopmental
that more work in this area is warranted. time windows such as adolescence. In addition, this highlights the
The emergence of maladaptive behavior is modeled in rodents importance of identifying and examining the various factors that
that have been exposed to early life adverse events, such as stress could be affecting the microbial profile of the adolescent gut.
or immune challenge. While it should be noted that many of the
studies in early immune challenge come from the autism litera- 6.2. Diet, alcohol and exercise – effects on the microbiota
ture, and that autism is a disease that is primarily diagnosed and
evident from early childhood onwards, this body of work pro- Nutrition, among other numerous environmental factors, has a
vides some excellent examples of studies worth examining that profound impact on both neurodevelopment and the gut micro-
show altered behaviour in adolescent rodents following early life biota (Innis, 2008; Skinner et al., 2010; Burokas et al., 2015).
immune challenge which have implications for other disorders Specifically, nutritional challenges during vulnerable developmen-
such as schizophrenia (Harvey and Boksa, 2012). Injecting preg- tal periods can have a detrimental impact on development and can
nant mice with valproic acid (VPA) results in pups developing induce functional and structural effects that persist throughout the
autistic-like behaviour, and intriguingly, altered social behavior is lifespan (Barker et al., 1993; Seckl and Meaney, 2004; Skinner et al.,
later observed in adolescent offspring accompanied by changes to 2010). Adequate nutrition is thus essential during adolescence in
commensal intestinal microbiota profiles (de Theije et al., 2014). order to support neurodevelopment and the establishment of the
Similar studies of maternal immune activation (MIA) injected preg- gut microbiota (Borre et al., 2014). Remarkable growth and devel-
nant dams with either lipopolysaccharide or propionic acid, and opment occur over the course of adolescence, and as such, there
also demonstrated significant changes to social behaviour in ado- is a great demand for calories and nutrients. In addition, adoles-
lescent offspring. Marked changes in enteric bacteria were also cence is characterized by an increased independence leading to
observed with both treatments (Foley et al., 2014). In a recent the establishment of food choices, which can highly augment the
study highlighting the potential role of intestinal bacteria in these risk of dietary deficiencies (Findlay, 2004) and excesses (Berall,
MIA-induced behavioural alterations, pregnant dams were injected 2002). More than ever before, adolescents have accessibility to a
with the viral mimic Poly (I:C), and then subsequent pups were huge amount of unhealthy food options. Westernization has driven
fed the human commensal Bacteroides fragilis. Fed pups later large modifications to dietary patterns with an increase in intake
showed attenuated changes to gut permeability, social behavior of fats, particularly saturated and trans-fats, and refined sugars
and anxiety-like behavior (Hsiao et al., 2013). Interestingly, MIA observed globally. This shift in diet has been linked to a loss of gut
treatment alters the expression of proteins associated with neu- microbial diversity including fiber-fermenting bacteria (De Filippo
ronal development and synaptic transmission in the pre-pubertal et al., 2010), and to the flourishing of a wide range of chronic dis-
rat offspring (Forrest et al., 2012). MIA offspring display reduced eases (Maslowski and Mackay, 2011). Diets high in fat and sugar
hippocampal myelination and axonal diameter in the juvenile, pre- have been shown to markedly alter the gut microbial composition
weaning stage, but not in adulthood (Makinodan et al., 2008). MIA by decreasing the Bacteroidetes/Firmicutes ratio and Lactobacillales
treatment also alters dendritic morphology in the prefrontal cor- numbers, which are associated with brain dysfunction and behav-
tex and hippocampus of offspring rats throughout development ioral changes (Pyndt Jorgensen et al., 2014; Bruce-Keller et al., 2015;
(Baharnoori et al., 2009). Similarly, prenatal VPA exposure induces Magnusson et al., 2015). Dietary fibre is known to be vital to shaping
neural remodelling in limbic regions in adult rats, but not in the of the gut microbiota (De Filippo et al., 2010; Schnorr et al., 2014),
peri-pubertal period (Bringas et al., 2013). and there has been a great reduction in fibre intake in the Western
diet. Interestingly, intake of a low-fibre diet over several genera-
6.1. Stress-resilience and microbiota tions has been demonstrated to result in a considerable loss of gut
microbial diversity, which could not be restored by a dietary fibre
In this review, we have discussed how the microbiota regulates re-intervention (Sonnenburg et al., 2016). A global dietary shift
the brain development and function, however, the inverse is also towards more Western-style foods has also led to a major decrease
true, and findings indicate that stressful experiences can in and of in intake of essential omega-3 polyunsaturated fatty acids (PUFAs)
themselves alter the composition of the gut microbiota (O’Mahony and an increase in omega-6 PUFAs (Simopoulos, 2011). A defi-
et al., 2009; Bailey et al., 2011; Dinan and Cryan, 2012; De Palma ciency in dietary n-3 PUFAs across consecutive generations of rats
et al., 2015; Golubeva et al., 2015; Jasarevic et al., 2015; Bharwani resulted in significant behavioural deficits during adolescence that
et al., 2016). These stressors can impact microbial composition not were linked to dopamine neurotransmission (Bondi et al., 2014),
only in early life but also in adulthood. Prenatal stress in mice alters which may be related to changes in gut microbiota composition.
the composition of the intestinal microbiota of the offspring along Several studies in mice have demonstrated the beneficial impact of
with gastrointestinal function, respiration, and stress responsiv- omega-3 PUFAs on gut microbiota (Yu et al., 2014; Kaliannan et al.,
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 307

2015), while high doses of omega-6 resulted in dysregulation of the fering from IBS (Niedzielin et al., 2001; Bauserman and Michail,
gut microbiota (Ghosh et al., 2013). Furthermore, supplementing 2005; Whorwell et al., 2006; Andriulli et al., 2008; Enck et al., 2008;
omega-3 PUFAs has been shown to restore the long-lasting micro- Enck et al., 2009; Hun, 2009; Williams et al., 2009; Francavilla et al.,
bial alterations induced by an early life stressor (Pusceddu et al., 2010; Guglielmetti et al., 2011; Clarke et al., 2012; Cui and Hu, 2012;
2015). Similarly, polyphenols, which are bioactive compounds nat- Dapoigny et al., 2012; Didari et al., 2015). However, other work has
urally found in plants, present a potential to modulate the gut shown that probiotics may also have beneficial effects on mood in
microbiota, thus showing prebiotic-like activity (Qiao et al., 2014; healthy volunteers (Benton et al., 2007; Messaoudi et al., 2011b;
Tzounis et al., 2011; Cardona et al., 2013; Kawabata et al., 2013). Messaoudi et al., 2011a; Mohammadi et al., 2015; Steenbergen
Some polyphenols have been shown to increase the growth of et al., 2015). Clearly the use of pre- and or probiotics in the treat-
Akkermansia muciniphila and decrease the proportion of Firmicutes ment of mood and mood associated disorders is a promising field,
to Bacteroidetes (Roopchand et al., 2015), as well as increase the and given these reports of improved symptoms in both clinical and
growth of both Lactobacillus and Bifidobacterium (Qiao et al., 2014), healthy populations, further study into bacterial-based therapeu-
all of which are considered to be beneficial changes to the host. tic interventions for psychiatric disease is warranted. As treatment
As previously mentioned, adolescence is the time period in options focused around commensal bacteria would be low risk and
which substance abuse and eating disorders typically first mani- with few, if any, side effects, this seems an ideal avenue of pur-
fest (Paus et al., 2008; O’Connor and Cryan, 2014). Indeed, alcohol suit for interventions particularly geared towards adolescents. In
is a substantial dietary disruptor of the gut microbiota, which is addition, the relative plasticity of the adolescent brain could make
particularly important during adolescence due to sometimes high it more receptive to neurogenic benefits conferred by potential
levels of consumption. Several studies have demonstrated the dis- psychobiotic therapy.
turbance of intestinal microbial homeostasis by the consumption of
alcohol in both rodents and humans (Mutlu et al., 2009; Mutlu et al.,
2012; Iyer and Vaishnava, 2016). Moreover, the microbiota profiles 8. Concluding remarks
of patients with the eating disorder anorexia nervosa show reduc-
tions in bacterial diversity (Kleiman et al., 2015). Interestingly, the The fact that the adolescence is a second, vital, critical window
levels of anxiety, depression, and eating disorder psychopathol- of brain development is something of a double-edged sword. On
ogy are associated with bacterial diversity (Kleiman et al., 2015). the one hand, the brain is arguably at its most vulnerable, second
Taken all together, it is apparent that diet is a key modulator only to the developing infant brain in terms of dramatic changes in
of the gut microbial composition, which can consequently influ- structuring, wiring and function that is occurring subject to external
ence brain function (Seira Oriach et al., 2016). It is therefore of influence from the individual’s environment. This shaping and re-
great importance to meet nutritional requirements during adoles- wiring is happening simultaneously with a time when the teenager
cence, as diet-induced changes in microbiota could contribute to is entering the world as an independent young adult, often fac-
the development of psychiatric disorders later in life. ing previously unknown risks and challenges. Alternately, this can
Recently, it has been shown that exercise is also an important be viewed as one of the most exciting periods of brain develop-
modulator of intestinal microbial status, particularly during the ment, an opportunity to harness the already naturally occurring
adolescent period. An intriguing study has shown that exposing developmental change underway. The adolescent window provides
juvenile rats to a running wheel for 6 weeks resulted in increases in an opening for the introduction of potential therapeutics, partic-
beneficial microbial species in the gut, with increased Bacteroidetes ularly those designed to address those psychiatric illnesses that
species and decreased Firmicutes observed. More alterations were frequently first appear in young adulthood. Indeed, there are many
observed in bacterial genera of adolescent rats first exposed to 6 examples of preventative measures directed at adolescents, those
weeks of exercise than adult rats exposed for the same period of designed in order to aid youth in navigating the various challenges
time. In addition, introducing exercise during the adolescent period they so frequently face. It would therefore be prudent to consider
resulted in a persistent increase in lean body mass, which was not the potential for therapeutics aimed at the intestinal microbiota
observed in the adult rat runners (Mika et al., 2015). The results of in the young adult. Individuals at high risk for the development
this study are provocative, and indicate that more work needs to of psychiatric disorders could be identified, and the potentially
be done in examining the effects of lifestyle choices on commen- preventative and/or ameliorative options of pre- and probiotics
sal bacteria and particularly those aimed at the adolescent period, could be considered as a safe and possibly extremely effective
as the opportunity for targeted interventions directed at intestinal way of improving teenage resiliency. The microbiota-gut-brain axis
microbiota could be substantial. presents an attractive target for the introduction of therapies that
may prove highly beneficial to the mental health of the developing
teen. As such, we need to turn our attention to the possibility that
7. Future directions microbial-based interventions could potentially improve teenage
mental resiliency. Given that we know the teenage years are ones in
Given our rapidly growing understanding of microbiota-gut- which the young adult often experiences difficult to control stress-
brain communication it is unsurprising that this axis is an attractive ors, accompanied by less than optimal diet and sleep patterns, pre-
target for the development of potential therapeutics for psychiatric and probiotic based therapies could provide an avenue by which
disease. Currently there is high global interest in the possibility of to bolster teenage resiliency. An increased understanding of ado-
using both and pre- and probiotics for the often refractory psy- lescent nervous system development in general, and the teenage
chiatric illnesses. “Psychobiotics” is the term proposed to refer microbiota-gut-brain axis in particular will aid us in identifying
to the therapeutic potential of manipulating the intestinal micro- exciting new treatment approaches for psychiatric illness in the
biota for the treatment of these diseases (Dinan et al., 2013). To young and may direct us to novel therapeutic channels for difficult
date, most of the clinical studies examining the potential bene- to treat mental illness throughout life.
fits of probiotics have focused on patients suffering from IBS. IBS
is a functional bowel disorder with symptoms of altered bowel
References
habits, visceral hypersensitivity and often co-morbid anxiety and
depressive symptoms. Several different strains of bacteria have Adlerberth, I., Wold, A.E., 2009. Establishment of the gut microbiota in Western
been demonstrated to have ameliorative effects in patients suf- infants. Acta Paediatr. 98, 229–238.
308 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

Agans, R., Rigsbee, L., Kenche, H., Michail, S., Khamis, H.J., Paliy, O., 2011. Distal gut Borre, Y.E., O’Keeffe, G.W., Clarke, G., Stanton, C., Dinan, T.G., Cryan, J.F., 2014.
microbiota of adolescent children is different from that of adults. FEMS Microbiota and neurodevelopmental windows: implications for brain
Microbiol. Ecol. 77, 404–412. disorders. Trends Mol. Med. 20, 509–518.
Andriulli, A., Neri, M., Loguercio, C., Terreni, N., Merla, A., Cardarella, M.P., Federico, Bourgeois, J.P., Rakic, P., 1993. Changes of synaptic density in the primary visual
A., Chilovi, F., Milandri, G.L., De Bona, M., Cavenati, S., Gullini, S., Abbiati, R., cortex of the macaque monkey from fetal to adult stage. J. Neurosci. 13,
Garbagna, N., Cerutti, R., Grossi, E., 2008. Clinical trial on the efficacy of a new 2801–2820.
symbiotic formulation, Flortec, in patients with irritable bowel syndrome: a Bourgeois, J.P., Goldman-Rakic, P.S., Rakic, P., 1994. Synaptogenesis in the
multicenter, randomized study. J. Clin. Gastroenterol. 42 (Suppl. 3), S218–S223 prefrontal cortex of rhesus monkeys. Cereb. Cortex 4, 78–96.
(Pt 2). Bravo, J.A., Forsythe, P., Chew, M.V., Escaravage, E., Savignac, H.M., Dinan, T.G.,
Arboleya, S., Sanchez, B., Milani, C., Duranti, S., Solis, G., Fernandez, N., de los Bienenstock, J., Cryan, J.F., 2011. Ingestion of Lactobacillus strain regulates
Reyes-Gavilan, C.G., Ventura, M., Margolles, A., Gueimonde, M., 2015. Intestinal emotional behavior and central GABA receptor expression in a mouse via the
microbiota development in preterm neonates and effect of perinatal vagus nerve. Proc. Natl. Acad. Sci. U. S. A. 108, 16050–16055.
antibiotics. J. Pediatr. 166, 538–544. Bringas, M.E., Carvajal-Flores, F.N., Lopez-Ramirez, T.A., Atzori, M., Flores, G., 2013.
Arentsen, T., Raith, H., Qian, Y., Forssberg, H., Diaz Heijtz, R., 2015. Host microbiota Rearrangement of the dendritic morphology in limbic regions and altered
modulates development of social preference in mice. Microb Ecol. Health Dis. exploratory behavior in a rat model of autism spectrum disorder. Neuroscience
26, 29719. 241, 170–187.
Azad, M.B., Konya, T., Maughan, H., Guttman, D.S., Field, C.J., Chari, R.S., Sears, M.R., Brizio, A., Gabbatore, I., Tirassa, M., Bosco, F.M., 2015. No more a child, not yet an
Becker, A.B., Scott, J.A., Kozyrskyj, A.L., 2013. Gut microbiota of healthy adult: studying social cognition in adolescence. Front. Psychol. 6, 1011.
Canadian infants: profiles by mode of delivery and infant diet at 4 months. Bruce-Keller, A.J., Salbaum, J.M., Luo, M., Blanchard Et, Taylor, C.M., Welsh, D.A.,
CMAJ 185, 385–394. Berthoud, H.R., 2015. Obese-type gut microbiota induce neurobehavioral
Backhed, F., Ley, R.E., Sonnenburg, J.L., Peterson, D.A., Gordon, J.I., 2005. changes in the absence of obesity. Biol. Psychiatry 77, 607–615.
Host-bacterial mutualism in the human intestine. Science 307, 1915–1920. Burnett, S., Sebastian, C., Cohen Kadosh, K., Blakemore, S.J., 2011. The social brain
Baharnoori, M., Brake, W.G., Srivastava, L.K., 2009. Prenatal immune challenge in adolescence: evidence from functional magnetic resonance imaging and
induces developmental changes in the morphology of pyramidal neurons of behavioural studies. Neurosci. Biobehav. Rev. 35, 1654–1664.
the prefrontal cortex and hippocampus in rats. Schizophr. Res. 107, 99–109. Burokas, A., Moloney, R.D., Dinan, T.G., Cryan, J.F., 2015. Microbiota regulation of
Bailey, M.T., Dowd, S.E., Galley, J.D., Hufnagle, A.R., Allen, R.G., Lyte, M., 2011. the Mammalian gut-brain axis. Adv. Appl. Microbiol. 91, 1–62.
Exposure to a social stressor alters the structure of the intestinal microbiota: Cairns, R.B., Cairns, B.D., Neckerman, H.J., Ferguson, L.L., Gariepy, J., 1989. Growth
implications for stressor-induced immunomodulation. Brain Behav. Immun. and aggression: 1. Childhood to early adolescence. Dev. Psychol. 25, 320–330.
25, 397–407. Cardona, F., Andres-Lacueva, C., Tulipani, S., Tinahones, F.J., Queipo-Ortuno, M.I.,
Barker, D.J., Gluckman, P.D., Godfrey, K.M., Harding, J.E., Owens, J.A., Robinson, J.S., 2013. Benefits of polyphenols on gut microbiota and implications in human
1993. Fetal nutrition and cardiovascular disease in adult life. Lancet 341, health. J. Nutr. Biochem. 24, 1415–1422.
938–941. Casey, B.J., Jones, R.M., Hare, T.A., 2008. The adolescent brain. Ann. N. Y. Acad. Sci.
Barnea-Goraly, N., Menon, V., Eckert, M., Tamm, L., Bammer, R., Karchemskiy, A., 1124, 111–126.
Dant, C.C., Reiss, A.L., 2005. White matter development during childhood and Cauffman, E., Shulman, E.P., Steinberg, L., Claus, E., Banich, M.T., Graham, S.,
adolescence: a cross-sectional diffusion tensor imaging study. Cereb. Cortex Woolard, J., 2010. Age differences in affective decision making as indexed by
15, 1848–1854. performance on the Iowa Gambling Task. Dev. Psychol. 46, 193–207.
Bauserman, M., Michail, S., 2005. The use of Lactobacillus GG in irritable bowel Clarke, G., Cryan, J.F., Dinan, T.G., Quigley, E.M., 2012. Review article: probiotics for
syndrome in children: a double-blind randomized control trial. J. Pediatr. 147, the treatment of irritable bowel syndrome – focus on lactic acid bacteria.
197–201. Aliment. Pharmacol. Ther. 35, 403–413.
Benes, F.M., 1989. Myelination of cortical-hippocampal relays during late Clarke, G., Grenham, S., Scully, P., Fitzgerald, P., Moloney, R.D., Shanahan, F., Dinan,
adolescence. Schizophr. Bull. 15, 585–593. T.G., Cryan, J.F., 2013. The microbiome-gut-brain axis during early life
Benton, D., Williams, C., Brown, A., 2007. Impact of consuming a milk drink regulates the hippocampal serotonergic system in a sex-dependent manner.
containing a probiotic on mood and cognition. Eur. J. Clin. Nutr. 61, 355–361. Mol. Psychiatry 18, 666–673.
Berall, G.B., 2002. Obesity: a crisis of growing proportions. Paediatr. Child Health 7, Cohen, J.R., Asarnow, R.F., Sabb, F.W., Bilder, R.M., Bookheimer, S.Y., Knowlton, B.J.,
325–328. Poldrack, R.A., 2010. A unique adolescent response to reward prediction errors.
Bercik, P., Verdu, E.F., Foster, J.A., Macri, J., Potter, M., Huang, X., Malinowski, P., Nat. Neurosci. 13, 669–671.
Jackson, W., Blennerhassett, P., Neufeld, K.A., Lu, J., Khan, W.I., Costa, M., Brookes, S.J., Hennig, G.W., 2000. Anatomy and physiology of the enteric
Corthesy-Theulaz, I., Cherbut, C., Bergonzelli, G.E., Collins, S.M., 2010. Chronic nervous system. Gut 47 (Suppl. 4), iv15–iv19.
gastrointestinal inflammation induces anxiety-like behavior and alters central Cragg, B.G., 1975. The development of synapses in the visual system of the cat. J.
nervous system biochemistry in mice. Gastroenterology 139, 2102–2112 Comp. Neurol. 160, 147–166.
(e2101). Crone, E.A., Dahl, R.E., 2012. Understanding adolescence as a period of
Bercik, P., Denou, E., Collins, J., Jackson, W., Lu, J., Jury, J., Deng, Y., Blennerhassett, social-affective engagement and goal flexibility. Nat. Rev. Neurosci. 13,
P., Macri, J., McCoy, K.D., Verdu, E.F., Collins, S.M., 2011a. The intestinal 636–650.
microbiota affect central levels of brain-derived neurotropic factor and Cryan, J.F., Dinan, T.G., 2012. Mind-altering microorganisms: the impact of the gut
behavior in mice. Gastroenterology 141, 599–609 (609 e591–593). microbiota on brain and behaviour. Nat. Rev. Neurosci. 13, 701–712.
Bercik, P., Park, A.J., Sinclair, D., Khoshdel, A., Lu, J., Huang, X., Deng, Y., Cryan, J.F., O’Mahony, S.M., 2011. The microbiome-gut-brain axis: from bowel to
Blennerhassett, P.A., Fahnestock, M., Moine, D., Berger, B., Huizinga, J.D., Kunze, behavior. Neurogastroenterol. Motil. 23, 187–192.
W., McLean, P.G., Bergonzelli, G.E., Collins, S.M., Verdu, E.F., 2011b. The Csikszentmihalyi, M., Larson, R., Prescott, S., 1977. The ecology of adolescent
anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways activity and experience. J. Youth Adolesc. 6, 281–294.
for gut-brain communication. Neurogastroenterol. Motil. 23, 1132–1139. Cui, S., Hu, Y., 2012. Multistrain probiotic preparation significantly reduces
Bercik, P., 2011. The microbiota-gut-brain axis: learning from intestinal bacteria? symptoms of irritable bowel syndrome in a double-blind placebo-controlled
Gut 60, 288–289. study. Int. J. Clin. Exp. Med. 5, 238–244.
Berndt, T.J., Keefe, K., 1979. Developmental changes in conformity to peers and Dahl, R.E., 2004. Adolescent brain development: a period of vulnerabilities and
parents. Dev. Psychol. 15, 608–616. opportunities. Keynote address. Ann. N. Y. Acad. Sci. 1021, 1–22.
Bezirtzoglou, E., Tsiotsias, A., Welling, G.W., 2011. Microbiota profile in feces of Dapoigny, M., Piche, T., Ducrotte, P., Lunaud, B., Cardot, J.M., Bernalier-Donadille,
breast- and formula-fed newborns by using fluorescence in situ hybridization A., 2012. Efficacy and safety profile of LCR35 complete freeze-dried culture in
(FISH). Anaerobe 17, 478–482. irritable bowel syndrome: a randomized, double-blind study. World J.
Bharwani, A., Mian, M.F., Foster, J.A., Surette, M.G., Bienenstock, J., Forsythe, P., Gastroenterol. 18, 2067–2075.
2016. Structural & functional consequences of chronic psychosocial stress on De Filippo, C., Cavalieri, D., Di Paola, M., Ramazzotti, M., Poullet, J.B., Massart, S.,
the microbiome & host. Psychoneuroendocrinology 63, 217–227. Collini, S., Pieraccini, G., Lionetti, P., 2010. Impact of diet in shaping gut
Bjork, J.M., Knutson, B., Fong, G.W., Caggiano, D.M., Bennett, S.M., Hommer, D.W., microbiota revealed by a comparative study in children from Europe and rural
2004. Incentive-elicited brain activation in adolescents: similarities and Africa. Proc. Natl. Acad. Sci. U. S. A. 107, 14691–14696.
differences from young adults. J. Neurosci. 24, 1793–1802. De Palma, G., Collins, S.M., Bercik, P., Verdu, E.F., 2014. The microbiota-gut-brain
Blackshaw, L.A., Brookes, S.J., Grundy, D., Schemann, M., 2007. Sensory axis in gastrointestinal disorders: stressed bugs, stressed brain or both? J.
transmission in the gastrointestinal tract. Neurogastroenterol. Motil. 19, 1–19. Physiol. 592, 2989–2997.
Blakemore, S.J., Choudhury, S., 2006. Development of the adolescent brain: De Palma, G., Blennerhassett, P., Lu, J., Deng, Y., Park, A.J., Green, W., Denou, E.,
implications for executive function and social cognition. J. Child Psychol. Silva, M.A., Santacruz, A., Sanz, Y., Surette, M.G., Verdu, E.F., Collins, S.M.,
Psychiatry 47, 296–312. Bercik, P., 2015. Microbiota and host determinants of behavioural phenotype
Blakemore, S.J., 2008. The social brain in adolescence. Nat. Rev. Neurosci. 9, in maternally separated mice. Nat. Commun. 6, 7735.
267–277. de Theije, C.G., Wopereis, H., Ramadan, M., van Eijndthoven, T., Lambert, J., Knol, J.,
Bondi, C.O., Taha, A.Y., Tock, J.L., Totah, N.K., Cheon, Y., Torres, G.E., Rapoport, S.I., Garssen, J., Kraneveld, A.D., Oozeer, R., 2014. Altered gut microbiota and
Moghaddam, B., 2014. Adolescent behavior and dopamine availability are activity in a murine model of autism spectrum disorders. Brain Behav. Immun.
uniquely sensitive to dietary omega-3 fatty acid deficiency. Biol. Psychiatry 75, 37, 197–206.
38–46. Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T.G., Cryan, J.F., 2014. Microbiota is
essential for social development in the mouse. Mol. Psychiatry 19, 146–148.
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 309

Desbonnet, L., Clarke, G., Traplin, A., O’Sullivan, O., Crispie, F., Moloney, R.D., Cotter, Furness, J.B., 2006. The organisation of the autonomic nervous system: peripheral
P.D., Dinan, T.G., Cryan, J.F., 2015. Gut microbiota depletion from early connections. Auton. Neurosci. 130, 1–5.
adolescence in mice: implications for brain and behaviour. Brain Behav. Galligan, J.J., Furness, J.B., Costa, M., 1989. Migration of the myoelectric complex
Immun. 48, 165–173. after interruption of the myenteric plexus: intestinal transection and
Diaz Heijtz, R., Wang, S., Anuar, F., Qian, Y., Bjorkholm, B., Samuelsson, A., Hibberd, regeneration of enteric nerves in the guinea pig. Gastroenterology 97,
M.L., Forssberg, H., Pettersson, S., 2011. Normal gut microbiota modulates 1135–1146.
brain development and behavior. Proc. Natl. Acad. Sci. U. S. A. 108, 3047–3052. Galvan, A., Hare, T.A., Parra, C.E., Penn, J., Voss, H., Glover, G., Casey, B.J., 2006.
Didari, T., Mozaffari, S., Nikfar, S., Abdollahi, M., 2015. Effectiveness of probiotics in Earlier development of the accumbens relative to orbitofrontal cortex might
irritable bowel syndrome: updated systematic review with meta-analysis. underlie risk-taking behavior in adolescents. J. Neurosci. 26, 6885–6892.
World J. Gastroenterol. 21, 3072–3084. Gardner, M., Steinberg, L., 2005. Peer influence on risk taking, risk preference, and
Dinan, T.G., Cryan, J.F., 2012. Regulation of the stress response by the gut risky decision making in adolescence and adulthood: an experimental study.
microbiota: implications for psychoneuroendocrinology. Dev. Psychol. 41, 625–635.
Psychoneuroendocrinology 37, 1369–1378. Gareau, M.G., Jury, J., MacQueen, G., Sherman, P.M., Perdue, M.H., 2007. Probiotic
Dinan, T.G., Stanton, C., Cryan, J.F., 2013. Psychobiotics: a novel class of treatment of rat pups normalises corticosterone release and ameliorates
psychotropic. Biol. Psychiatry 74, 720–726. colonic dysfunction induced by maternal separation. Gut 56, 1522–1528.
Dominguez-Bello, M.G., Costello, E.K., Contreras, M., Magris, M., Hidalgo, G., Fierer, Gareau, M.G., Wine, E., Rodrigues, D.M., Cho, J.H., Whary, M.T., Philpott, D.J.,
N., Knight, R., 2010. Delivery mode shapes the acquisition and structure of the Macqueen, G., Sherman, P.M., 2011. Bacterial infection causes stress-induced
initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. memory dysfunction in mice. Gut 60, 307–317.
Sci. U. S. A. 107, 11971–11975. Ghosh, S., DeCoffe, D., Brown, K., Rajendiran, E., Estaki, M., Dai, C., Yip, A., Gibson,
Dominguez-Bello, M.G., Blaser, M.J., Ley, R.E., Knight, R., 2011. Development of the D.L., 2013. Fish oil attenuates omega-6 polyunsaturated fatty acid-induced
human gastrointestinal microbiota and insights from high-throughput dysbiosis and infectious colitis but impairs LPS dephosphorylation activity
sequencing. Gastroenterology 140, 1713–1719. causing sepsis. PLoS One 8, e55468.
Eiland, L., Romeo, R.D., 2013. Stress and the developing adolescent brain. Giaroni, C., De Ponti, F., Cosentino, M., Lecchini, S., Frigo, G., 1999. Plasticity in the
Neuroscience 249, 162–171. enteric nervous system. Gastroenterology 117, 1438–1458.
El Aidy, S., Dinan, T.G., Cryan, J.F., 2015. Gut microbiota: the conductor in the Gibson, G.R., Roberfroid, M.B., 1995. Dietary modulation of the human colonic
orchestra of immune-neuroendocrine communication. Clin. Ther. 37, 954–967. microbiota: introducing the concept of prebiotics. J. Nutr. 125 (6), 1401–1412.
Enck, P., Zimmermann, K., Menke, G., Muller-Lissner, S., Martens, U., Klosterhalfen, Giedd, J.N., Snell, J.W., Lange, N., Rajapakse, J.C., Casey, B.J., Kozuch, P.L., Vaituzis,
S., 2008. A mixture of Escherichia coli (DSM 17252) and Enterococcus faecalis A.C., Vauss, Y.C., Hamburger, S.D., Kaysen, D., Rapoport, J.L., 1996. Quantitative
(DSM 16440) for treatment of the irritable bowel syndrome – a randomized magnetic resonance imaging of human brain development: ages 4–18. Cereb.
controlled trial with primary care physicians. Neurogastroenterol. Motil. 20, Cortex 6, 551–560.
1103–1109. Giedd, J.N., Blumenthal, J., Jeffries, N.O., Castellanos, F.X., Liu, H., Zijdenbos, A., Paus,
Enck, P., Zimmermann, K., Menke, G., Klosterhalfen, S., 2009. Randomized T., Evans, A.C., Rapoport, J.L., 1999. Brain development during childhood and
controlled treatment trial of irritable bowel syndrome with a probiotic E.-coli adolescence: a longitudinal MRI study. Nat. Neurosci. 2, 861–863.
preparation (DSM17252) compared to placebo. Z Gastroenterol 47, 209–214. Giedd, J.N., 2009. Linking adolescent sleep, brain maturation, and behavior. J.
Erny, D., Hrabe de Angelis, A.L., Jaitin, D., Wieghofer, P., Staszewski, O., David, E., Adolesc. Health 45, 319–320.
Keren-Shaul, H., Mahlakoiv, T., Jakobshagen, K., Buch, T., Schwierzeck, V., Giorgio, A., Watkins, K.E., Chadwick, M., James, S., Winmill, L., Douaud, G., De
Utermohlen, O., Chun, E., Garrett, W.S., McCoy, K.D., Diefenbach, A., Staeheli, P., Stefano, N., Matthews, P.M., Smith, S.M., Johansen-Berg, H., James, A.C., 2010.
Stecher, B., Amit, I., Prinz, M., 2015. Host microbiota constantly control Longitudinal changes in grey and white matter during adolescence.
maturation and function of microglia in the CNS. Nat. Neurosci. 18, 965–977. Neuroimage 49, 94–103.
Fair, D.A., Cohen, A.L., Dosenbach, N.U., Church, J.A., Miezin, F.M., Barch, D.M., Goddings, A.L., Mills, K.L., Clasen, L.S., Giedd, J.N., Viner, R.M., Blakemore, S.J., 2014.
Raichle, M.E., Petersen, S.E., Schlaggar, B.L., 2008. The maturing architecture of The influence of puberty on subcortical brain development. Neuroimage 88,
the brain’s default network. Proc. Natl. Acad. Sci. U. S. A. 105, 4028–4032. 242–251.
Fallani, M., Young, D., Scott, J., Norin, E., Amarri, S., Adam, R., Aguilera, M., Khanna, Goehler, L.E., Park, S.M., Opitz, N., Lyte, M., Gaykema, R.P., 2008. Campylobacter
S., Gil, A., Edwards, C.A., Dore, J., 2010. Intestinal microbiota of 6-week-old jejuni infection increases anxiety-like behavior in the holeboard: possible
infants across Europe: geographic influence beyond delivery mode, anatomical substrates for viscerosensory modulation of exploratory behavior.
breast-feeding, and antibiotics. J. Pediatr. Gastroenterol. Nutr. 51, 77–84. Brain Behav. Immun. 22, 354–366.
Falony, G., Joossens, M., Vieira-Silva, S., Wang, J., Darzi, Y., Faust, K., Kurilshikov, A., Goldman, L., Winget, C., Hollingshead, G.W., Levine, S., 1973. Postweaning
Bonder, M.J., Valles-Colomer, M., Vandeputte, D., Tito, R.Y., Chaffron, S., development of negative feedback in the pituitary-adrenal system of the rat.
Rymenans, L., Verspecht, C., De Sutter, L., Lima-Mendez, G., D’hoe, K., Neuroendocrinology 12, 199–211.
Jonckheere, K., Homola, D., Garcia, R., Tigchelaar, E.F., Eeckhaudt, L., Fu, J., Golubeva, A.V., Crampton, S., Desbonnet, L., Edge, D., O’Sullivan, O., Lomasney,
Henckaerts, L., Zhernakova, A., Wijmenga, C., Raes, J., 2016. Population-level K.W., Zhdanov, A.V., Crispie, F., Moloney, R.D., Borre, Y.E., Cotter, P.D., Hyland,
analysis of gut microbiome variation. Science 352 (6285), 560–564. N.P., O’Halloran, K.D., Dinan, T.G., O’Keeffe, G.W., Cryan, J.F., 2015. Prenatal
Findlay, S.M., 2004. Dieting in adolescence. Paediatr. Child Health 9, 487–491. stress-induced alterations in major physiological systems correlate with gut
Flint, H.J., Scott, K.P., Duncan, S.H., Louis, P., Forano, E., 2012. Microbial degradation microbiota composition in adulthood. Psychoneuroendocrinology 60, 58–74.
of complex carbohydrates in the gut. Gut Microbes 3 (4), 289–306. Guglielmetti, S., Mora, D., Gschwender, M., Popp, K., 2011. Randomised clinical
Foley, K.A., MacFabe, D.F., Vaz, A., Ossenkopp, K.P., Kavaliers, M., 2014. Sexually trial: bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel
dimorphic effects of prenatal exposure to propionic acid and syndrome and improves quality of life?a double-blind, placebo-controlled
lipopolysaccharide on social behavior in neonatal, adolescent, and adult rats: study. Aliment. Pharmacol. Ther. 33, 1123–1132.
implications for autism spectrum disorders. Int. J. Dev. Neurosci. 39, 68–78. Hansen, T.H., Gobel, R.J., Hansen, T., Pedersen, O., 2015. The gut microbiome in
Forde, B.M., O’Toole, P.W., 2013. Next-generation sequencing technologies and cardio-metabolic health. Genome Med. 7, 33.
their impact on microbial genomics. Briefings Funct. Genomics 12, 440–453. Hare, T.A., Tottenham, N., Galvan, A., Voss, H.U., Glover, G.H., Casey, B.J., 2008.
Forrest, C.M., Khalil, O.S., Pisar, M., Smith, R.A., Darlington, L.G., Stone, T.W., 2012. Biological substrates of emotional reactivity and regulation in adolescence
Prenatal activation of Toll-like receptors-3 by administration of the viral during an emotional go-nogo task. Biol. Psychiatry 63, 927–934.
mimetic poly(I:C) changes synaptic proteins, N-methyl-D-aspartate receptors Harvey, L., Boksa, P., 2012. Prenatal and postnatal animal models of immune
and neurogenesis markers in offspring. Mol. Brain 5, 22. activation: relevance to a range of neurodevelopmental disorders. Dev.
Foster, J.A., McVey Neufeld, K.A., 2013. Gut-brain axis: how the microbiome Neurobiol. 72 (10), 1335–1348.
influences anxiety and depression. Trends Neurosci. 36, 305–312. Hoban, A.E., Stilling, R.M., Ryan, F.J., Claesson, M.J., Shanahan, F., Dinan, T.G., Clarke,
Fouhy, F., Ross, R.P., Fitzgerald, G.F., Stanton, C., Cotter, P.D., 2012. Composition of G., Cryan, J.F., 2016. Regulation of prefrontal cortex myelination by the
the early intestinal microbiota: knowledge, knowledge gaps and the use of microbiota. Transl. Psychiatry 6, e774, http://dx.doi.org/10.1038/tp.2016.42.
high-throughput sequencing to address these gaps. Gut Microbes 3, 203–220. Hollister, E.B., Riehle, K., Luna, R.A., Weidler, E.M., Rubio-Gonzales, M., Mistretta,
Francavilla, R., Miniello, V., Magista, A.M., De Canio, A., Bucci, N., Gagliardi, F., T.A., Raza, S., Doddapaneni, H.V., Metcalf, G.A., Muzny, D.M., Gibbs, R.A.,
Lionetti, E., Castellaneta, S., Polimeno, L., Peccarisi, L., Indrio, F., Cavallo, L., Petrosino, J.F., Shulman, R.J., Versalovic, J., 2015. Structure and function of the
2010. A randomized controlled trial of Lactobacillus GG in children with healthy pre-adolescent pediatric gut microbiome. Microbiome 3, 36.
functional abdominal pain. Pediatrics 126, e1445–e1452. Holtmaat, A., Svoboda, K., 2009. Experience-dependent structural synaptic
Frank, D.N., Pace, N.R., 2008. Gastrointestinal microbiology enters the plasticity in the mammalian brain. Nat. Rev. Neurosci. 10, 647–658.
metagenomics era. Curr. Opin. Gastroenterol. 24, 4–10. Hopkins, M.J., Sharp, R., Macfarlane, G.T., 2002. Variation in human intestinal
Frith, U., Frith, C.D., 2003. Development and neurophysiology of mentalizing. microbiota with age. Digestive Liver Dis. 34 (Suppl. 2), S12–S18.
Philos. Trans. R. Soc. Lond. B Biol. Sci. 358, 459–473. Howitt, M.R., Garrett, W.S., 2012. A complex microworld in the gut: gut microbiota
Frohlich, E.E., Farzi, A., Mayerhofer, R., Reichmann, F., Jacan, A., Wagner, B., Zinser, and cardiovascular disease connectivity. Nat. Med. 18, 1188–1189.
E., Bordag, N., Magnes, C., Frohlich, E., Kashofer, K., Gorkiewicz, G., Holzer, P., Hsiao, E.Y., McBride, S.W., Hsien, S., Sharon, G., Hyde, E.R., McCue, T., Codelli, J.A.,
2016. Cognitive impairment by antibiotic-induced gut dysbiosis: analysis of Chow, J., Reisman, S.E., Petrosino, J.F., Patterson, P.H., Mazmanian, S.K., 2013.
gut microbiota-brain communication. Brain Behav. Immun., http://dx.doi.org/ Microbiota modulate behavioral and physiological abnormalities associated
10.1016/j.bbi.2016.02.020, pii: S0889-1591(16)30040-X. with neurodevelopmental disorders. Cell 155, 1451–1463.
Funkhouser, L.J., Bordenstein, S.R., 2013. Mom knows best: the universality of Hua, J.Y., Smith, S.J., 2004. Neural activity and the dynamics of central nervous
maternal microbial transmission. PLoS Biol. 11, e1001631. system development. Nat. Neurosci. 7, 327–332.
310 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

Hun, L., 2009. Bacillus coagulans significantly improved abdominal pain and Makinodan, M., Tatsumi, K., Manabe, T., Yamauchi, T., Makinodan, E., Matsuyoshi,
bloating in patients with IBS. Postgrad. Med. 121, 119–124. H., Shimoda, S., Noriyama, Y., Kishimoto, T., Wanaka, A., 2008. Maternal
Hussey, S., Wall, R., Gruffman, E., O’Sullivan, L., Ryan, C.A., Murphy, B., Fitzgerald, immune activation in mice delays myelination and axonal development in the
G., Stanton, C., Ross, R.P., 2011. Parenteral antibiotics reduce bifidobacteria hippocampus of the offspring. J. Neurosci. Res. 86, 2190–2200.
colonization and diversity in neonates. Int. J. Microbiol., 2011. Marceau, K., Ruttle, P.L., Shirtcliff, E.A., Essex, M.J., Susman, E.J., 2015.
Huttenlocher, P.R., Dabholkar, A.S., 1997. Regional differences in synaptogenesis in Developmental and contextual considerations for adrenal and gonadal
human cerebral cortex. J. Comp. Neurol. 387, 167–178. hormone functioning during adolescence: implications for adolescent mental
Huttenlocher, P.R., de Courten, C., Garey, L.J., Van der Loos, H., 1982. health. Dev. Psychobiol. 57, 742–768.
Synaptogenesis in human visual cortex?evidence for synapse elimination Markham, J.A., Greenough, W.T., 2004. Experience-driven brain plasticity: beyond
during normal development. Neurosci. Lett. 33, 247–252. the synapse. Neuron Glia Biol. 1, 351–363.
Huttenlocher, P.R., 1979. Synaptic density in human frontal cortex – Markle, J.G.M., Frank, D.N., Mortin-Toth, S., Robertson, C.E., Feazel, L.M.,
developmental changes and effects of aging. Brain Res. 163, 195–205. Rolle-Kampczyk, U., von Bergen, M., McCoy, K.D., Macpherson, A.J., Danska, J.S.,
Innis, S.M., 2008. Dietary omega 3 fatty acids and the developing brain. Brain Res. 2013. Sex differences in the gut microbiome drive hormone-dependent
1237, 35–43. regulation of autoimmunity. Science 339, 1084–1088 (80-).
Iyer, N., Vaishnava, S., 2016. Alcohol lowers your (Intestinal) inhibitions. Cell Host Maslowski, K.M., Mackay, C.R., 2011. Diet, gut microbiota and immune responses.
Microbe 19, 131–133. Nat. Immunol. 12, 5–9.
Jasarevic, E., Howerton, C.L., Howard, C.D., Bale, T.L., 2015. Alterations in the Mayer, E.A., Tillisch, K., Gupta, A., 2015. Gut/brain axis and the microbiota. J. Clin.
vaginal microbiome by maternal stress are associated with metabolic Invest. 125, 926–938.
reprogramming of the offspring gut and brain. Endocrinology 156, 3265–3276. Mayer, E.A., 2000. The neurobiology of stress and gastrointestinal disease. Gut 47,
Jasarevic, E., Morrison, K.E., Bale, T.L., 2016. Sex differences in the gut 861–869.
microbiome-brain axis across the lifespan. Philos. Trans. R. Soc. Lond. B Biol. McCormick, C.M., Mathews, I.Z., 2007. HPA function in adolescence: role of sex
Sci., 371. hormones in its regulation and the enduring consequences of exposure to
Jaworska, N., MacQueen, G., 2015. Adolescence as a unique developmental period. stressors. Pharmacol. Biochem. Behav. 86, 220–233.
J. Psychiatry Neurosci. 40, 291–293. McCormick, C.M., Smith, C., Mathews, I.Z., 2008. Effects of chronic social stress in
Jeffery, I.B., O’Toole, P.W., Ohman, L., Claesson, M.J., Deane, J., Quigley, E.M., Simren, adolescence on anxiety and neuroendocrine response to mild stress in male
M., 2012. An irritable bowel syndrome subtype defined by species-specific and female rats. Behav. Brain Res. 187, 228–238.
alterations in faecal microbiota. Gut 61, 997–1006. McVey Neufeld, K.A., Mao, Y.K., Bienenstock, J., Foster, J.A., Kunze, W.A., 2013. The
Juraska, J.M., Sisk, C.L., DonCarlos, L.L., 2013. Sexual differentiation of the microbiome is essential for normal gut intrinsic primary afferent neuron
adolescent rodent brain: hormonal influences and developmental excitability in the mouse. Neurogastroenterol. Motil. 25, 183–e188.
mechanisms. Horm. Behav. 64, 203–210. McVey Neufeld, K.A., Perez-Burgos, A., Mao, Y.K., Bienenstock, J., Kunze, W.A., 2015.
Kaliannan, K., Wang, B., Li, X.Y., Kim, K.J., Kang, J.X., 2015. A host-microbiome The gut microbiome restores intrinsic and extrinsic nerve function in
interaction mediates the opposing effects of omega-6 and omega-3 fatty acids germ-free mice accompanied by changes in calbindin. Neurogastroenterol.
on metabolic endotoxemia. Sci. Rep. 5, 11276. Motil. 27, 627–636.
Karlsson, F.H., Fak, F., Nookaew, I., Tremaroli, V., Fagerberg, B., Petranovic, D., McVey Neufeld, K.A., Luczynski, P., Dinan, T.G., Cryan, J.F., 2016. Reframing the
Backhed, F., Nielsen, J., 2012. Symptomatic atherosclerosis is associated with teenage wasteland: adolescent microbiota-gut-brain axis. Can. J. Psychiatry 61
an altered gut metagenome. Nat. Commun. 3, 1245. (4), http://dx.doi.org/10.1177/0706743716635536, 214–21.
Karlsson, F.H., Tremaroli, V., Nookaew, I., Bergstrom, G., Behre, C.J., Fagerberg, B., Messaoudi, M., Violle, N., Bisson, J.F., Desor, D., Javelot, H., Rougeot, C., 2011a.
Nielsen, J., Backhed, F., 2013. Gut metagenome in European women with Beneficial psychological effects of a probiotic formulation (Lactobacillus
normal, impaired and diabetic glucose control. Nature 498, 99–103. helveticus R0052 and Bifidobacterium longum R0175) in healthy human
Kawabata, K., Sugiyama, Y., Sakano, T., Ohigashi, H., 2013. Flavonols enhanced volunteers. Gut. Microbes 2, 256–261.
production of anti-inflammatory substance(s) by Bifidobacterium adolescentis: Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D., Nejdi, A., Bisson, J.F.,
prebiotic actions of galangin, quercetin, and fisetin. Biofactors 39, 422–429. Rougeot, C., Pichelin, M., Cazaubiel, M., Cazaubiel, J.M., 2011b. Assessment of
Kessler, R.C., Berglund, P., Demler, O., Jin, R., Merikangas, K.R., Walters, E.E., 2005. psychotropic-like properties of a probiotic formulation (Lactobacillus
Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the helveticus R0052 and Bifidobacterium longum R0175) in rats and human
National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62, 593–602. subjects. Br. J. Nutr. 105, 755–764.
Kilford, E., Garrett, E., Blakemore, S.-J., 2016. The development of social cognition Mika, A., Van Treuren, W., Gonzalez, A., Herrera, J.J., Knight, R., Fleshner, M., 2015.
in adolescence: an integrated perspective. In Special Issue on the Adolescent Exercise is more effective at altering gut microbial composition and producing
Brain. Neurosci. Biobehav. Rev. 70, 106–120. stable changes in lean mass in juvenile versus adult male F344 rats. PLoS One
Kleiman, S.C., Watson, H.J., Bulik-Sullivan, E.C., Huh, E.Y., Tarantino, L.M., Bulik, 10, e0125889.
C.M., Carroll, I.M., 2015. The intestinal microbiota in acute anorexia nervosa Miller, E.K., Cohen, J.D., 2001. An integrative theory of prefrontal cortex function.
and during renourishment: relationship to depression, anxiety, and eating Annu. Rev. Neurosci. 24, 167–202.
disorder psychopathology. Psychosom. Med. 77, 969–981. Mohammadi, A.A., Jazayeri, S., Khosravi-Darani, K., Solati, Z., Mohammadpour, N.,
Koren, O., Spor, A., Felin, J., Fak, F., Stombaugh, J., Tremaroli, V., Behre, C.J., Knight, Asemi, Z., Adab, Z., Djalali, M., Tehrani-Doost, M., Hosseini, M., Eghtesadi, S.,
R., Fagerberg, B., Ley, R.E., Backhed, F., 2011. Human oral, gut, and plaque 2015. The effects of probiotics on mental health and
microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. U. S. A. 108 hypothalamic-pituitary-adrenal axis: a randomized, double-blind,
(Suppl. 1), 4592–4598. placebo-controlled trial in petrochemical workers. Nutr. Neurosci., http://dx.
Kunze, W.A., Mao, Y.K., Wang, B., Huizinga, J.D., Ma, X., Forsythe, P., Bienenstock, J., doi.org/10.1179/1476830515Y.0000000023.
2009. Lactobacillus reuteri enhances excitability of colonic AH neurons by Monk, C.S., McClure, E.B., Nelson, E.E., Zarahn, E., Bilder, R.M., Leibenluft, E.,
inhibiting calcium-dependent potassium channel opening. J. Cell. Mol. Med. Charney, D.S., Ernst, M., Pine, D.S., 2003. Adolescent immaturity in
13, 2261–2270. attention-related brain engagement to emotional facial expressions.
Le Huerou-Luron, I., Blat, S., Boudry, G., 2010. Breast- v. formula-feeding: impacts Neuroimage 20, 420–428.
on the digestive tract and immediate and long-term health effects. Nutr. Res. Mueller, N.T., Whyatt, R., Hoepner, L., Oberfield, S., Dominguez-Bello, M.G., Widen,
Rev. 23, 23–36. E.M., Hassoun, A., Perera, F., Rundle, A., 2015. Prenatal exposure to antibiotics,
LeDoux, J., 2007. The amygdala. Curr. Biol. 17, R868–R874. cesarean section and risk of childhood obesity. Int. J. Obes. (Lond.) 39, 665–670.
Leussis, M.P., Andersen, S.L., 2008. Is adolescence a sensitive period for depression? Mutlu, E., Keshavarzian, A., Engen, P., Forsyth, C.B., Sikaroodi, M., Gillevet, P., 2009.
Behavioral and neuroanatomical findings from a social stress model. Synapse Intestinal dysbiosis: a possible mechanism of alcohol-induced endotoxemia
62, 22–30. and alcoholic steatohepatitis in rats. Alcohol Clin. Exp. Res. 33, 1836–1846.
Ley, R.E., Turnbaugh, P.J., Klein, S., Gordon, J.I., 2006. Microbial ecology: human gut Mutlu, E.A., Gillevet, P.M., Rangwala, H., Sikaroodi, M., Naqvi, A., Engen, P.A.,
microbes associated with obesity. Nature 444, 1022–1023. Kwasny, M., Lau, C.K., Keshavarzian, A., 2012. Colonic microbiome is altered in
Liston, C., McEwen, B.S., Casey, B.J., 2009. Psychosocial stress reversibly disrupts alcoholism. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G966–G978.
prefrontal processing and attentional control. Proc. Natl. Acad. Sci. U. S. A. 106, Nesse, R.M., Berridge, K.C., 1997. Psychoactive drug use in evolutionary
912–917. perspective. Science 278, 63–66.
Luczynski, P., McVey Neufeld, K.A., Seira Oriach, C., Clarke, G., Dinan, T.G., Cryan, J.F., Neufeld, K.M., Kang, N., Bienenstock, J., Foster, J.A., 2011. Reduced anxiety-like
2016. Growing up in a Bubble: using germ-free animals to assess the influence behavior and central neurochemical change in germ-free mice.
of the gut microbiota on brain and behaviour. Int. J. Neuropsychopharmacol. Neurogastroenterol. Motil. 23, 255–264 (e119).
Lyte, M., Varcoe, J.J., Bailey, M.T., 1998. Anxiogenic effect of subclinical bacterial Niedzielin, K., Kordecki, H., Birkenfeld, B., 2001. A controlled, double-blind,
infection in mice in the absence of overt immune activation. Physiol. Behav. 65, randomized study on the efficacy of Lactobacillus plantarum 299 V in patients
63–68. with irritable bowel syndrome. Eur. J. Gastroenterol. Hepatol. 13, 1143–1147.
Lyte, M., Li, W., Opitz, N., Gaykema, R.P., Goehler, L.E., 2006. Induction of O’Connor, R.M., Cryan, J.F., 2014. Adolescent brain vulnerability and
anxiety-like behavior in mice during the initial stages of infection with the psychopathology through the generations: role of diet and dopamine. Biol.
agent of murine colonic hyperplasia Citrobacter rodentium. Physiol. Behav. 89, Psychiatry 75, 4–6.
350–357. O’Mahony, S.M., Marchesi, J.R., Scully, P., Codling, C., Ceolho, A.M., Quigley, E.M.,
Magnusson, K.R., Hauck, L., Jeffrey, B.M., Elias, V., Humphrey, A., Nath, R., Perrone, Cryan, J.F., Dinan, T.G., 2009. Early life stress alters behavior, immunity, and
A., Bermudez, L.E., 2015. Relationships between diet-related changes in the gut microbiota in rats: implications for irritable bowel syndrome and psychiatric
microbiome and cognitive flexibility. Neuroscience 300, 128–140. illnesses. Biol. Psychiatry 65, 263–267.
K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312 311

Olson, E.A., Hooper, C.J., Collins, P., Luciana, M., 2007. Adolescents’ performance on Salonen, A., de Vos, W.M., Palva, A., 2010. Gastrointestinal microbiota in irritable
delay and probability discounting tasks: contributions of age, intelligence, bowel syndrome: present state and perspectives. Microbiology 156,
executive functioning, and self-reported externalizing behavior. Personality 3205–3215.
Individ. Differences 43, 1886–1897. Savignac, H.M., Tramullas, M., Kiely, B., Dinan, T.G., Cryan, J.F., 2015. Bifidobacteria
Paus, T., Keshavan, M., Giedd, J.N., 2008. Why do many psychiatric disorders modulate cognitive processes in an anxious mouse strain. Behav. Brain Res.
emerge during adolescence? Nat. Rev. Neurosci. 9, 947–957. 287, 59–72.
Penders, J., Thijs, C., Vink, C., Stelma, F.F., Snijders, B., Kummeling, I., van den Schafer, D.P., Lehrman, E.K., Kautzman, A.G., Koyama, R., Mardinly, A.R., Yamasaki,
Brandt, P.A., Stobberingh, E.E., 2006. Factors influencing the composition of the R., Ransohoff, R.M., Greenberg, M.E., Barres, B.A., Stevens, B., 2012. Microglia
intestinal microbiota in early infancy. Pediatrics 118, 511–521. sculpt postnatal neural circuits in an activity and complement-dependent
Perez-Burgos, A., Wang, B., Mao, Y.K., Mistry, B., McVey Neufeld, K.A., Bienenstock, manner. Neuron 74, 691–705.
J., Kunze, W., 2013. Psychoactive bacteria Lactobacillus rhamnosus (JB-1) Schnorr, S.L., Candela, M., Rampelli, S., Centanni, M., Consolandi, C., Basaglia, G.,
elicits rapid frequency facilitation in vagal afferents. Am. J. Physiol. Turroni, S., Biagi, E., Peano, C., Severgnini, M., Fiori, J., Gotti, R., De Bellis, G.,
Gastrointest. Liver Physiol. 304, G211–G220. Luiselli, D., Brigidi, P., Mabulla, A., Marlowe, F., Henry, A.G., Crittenden, A.N.,
Perez-Burgos, A., Mao, Y.K., Bienenstock, J., Kunze, W.A., 2014. The gut-brain axis 2014. Gut microbiome of the Hadza hunter-gatherers. Nat. Commun. 5, 3654.
rewired: adding a functional vagal nicotinic sensory synapse. FASEB J. 28, Seckl, J.R., Meaney, M.J., 2004. Glucocorticoid programming. Ann. N. Y. Acad. Sci.
3064–3074. 1032, 63–84.
Pfefferbaum, B., Wood, P.B., 1994. Self-report study of impulsive and delinquent Seira Oriach, C., Robertson, R.C., Stanton, C., Cryan, J.F., Dinan, T.G., 2016. Food for
behavior in college students. J. Adolesc. Health 15, 295–302. thought: the role of nutrition in the microbiota-gut-brain axis. Clin. Nutr. Exp.
Pfefferbaum, A., Mathalon, D.H., Sullivan, E.V., Rawles, J.M., Zipursky, R.B., Lim, K.O., 6, 25–38, http://dx.doi.org/10.1016/j.yclnex.2016.01.003.
1994. A quantitative magnetic resonance imaging study of changes in brain Sekar, A., Bialas, A.R., de Rivera, H., Davis, A., Hammond, T.R., Kamitaki, N., Tooley,
morphology from infancy to late adulthood. Arch. Neurol. 51, 874–887. K., Presumey, J., Baum, M., Van Doren, V., Genovese, G., Rose, S.A., Handsaker,
Pignatelli, D., Xiao, F., Gouveia, A.M., Ferreira, J.G., Vinson, G.P., 2006. Adrenarche in R.E., Daly, M.J., Carroll, M.C., Stevens, B., McCarroll, S.A., 2016. Schizophrenia
the rat. J. Endocrinol. 191, 301–308. risk from complex variation of complement component 4. Nature 530,
Pine, D.S., Grun, J., Zarahn, E., Fyer, A., Koda, V., Li, W., Szeszko, P.R., Ardekani, B., 177–183.
Bilder, R.M., 2001. Cortical brain regions engaged by masked emotional faces in Simopoulos, A.P., 2011. Evolutionary aspects of diet: the omega-6/omega-3 ratio
adolescents and adults: an fMRI study. Emotion 1, 137–147. and the brain. Mol. Neurobiol. 44, 203–215.
Power, S.E., O’Toole, P.W., Stanton, C., Ross, R.P., Fitzgerald, G.F., 2014. Intestinal Skinner, M.K., Manikkam, M., Guerrero-Bosagna, C., 2010. Epigenetic
microbiota, diet and health. Br. J. Nutr. 111 (3), 387–402. transgenerational actions of environmental factors in disease etiology. Trends
Prince, A.L., Antony, K.M., Chu, D.M., Aagaard, K.M., 2014. The microbiome, Endocrinol. Metab. 21, 214–222.
parturition, and timing of birth: more questions than answers. J. Reprod. Smith, C.J., Emge, J.R., Berzins, K., Lung, L., Khamishon, R., Shah, P., Rodrigues, D.M.,
Immunol. 104–105, 12–19. Sousa, A.J., Reardon, C., Sherman, P.M., Barrett, K.E., Gareau, M.G., 2014.
Pusceddu, M.M., El Aidy, S., Crispie, F., O’Sullivan, O., Cotter, P., Stanton, C., Kelly, P., Probiotics normalize the gut-brain-microbiota axis in immunodeficient mice.
Cryan, J.F., Dinan, T.G., 2015. N-3 polyunsaturated fatty acids (PUFAs) reverse Am. J. Physiol. Gastrointest. Liver Physiol. 307, G793–G802.
the impact of early-Life stress on the gut microbiota. PLoS One 10, e0139721. Sonnenburg, E.D., Smits, S.A., Tikhonov, M., Higginbottom, S.K., Wingreen, N.S.,
Pyndt Jorgensen, B., Hansen, J.T., Krych, L., Larsen, C., Klein, A.B., Nielsen, D.S., Sonnenburg, J.L., 2016. Diet-induced extinctions in the gut microbiota
Josefsen, K., Hansen, A.K., Sorensen, D.B., 2014. A possible link between food compound over generations. Nature 529, 212–215.
and mood: dietary impact on gut microbiota and behavior in BALB/c mice. Sowell, E.R., Thompson, P.M., Tessner, K.D., Toga, A.W., 2001. Mapping continued
PLoS One 9, e103398. brain growth and gray matter density reduction in dorsal frontal cortex:
Qiao, Y., Sun, J., Xia, S., Tang, X., Shi, Y., Le, G., 2014. Effects of resveratrol on gut inverse relationships during postadolescent brain maturation. J. Neurosci. 21,
microbiota and fat storage in a mouse model with high-fat-induced obesity. 8819–8829.
Food Funct. 5, 1241–1249. Sowell, E.R., Peterson, B.S., Thompson, P.M., Welcome, S.E., Henkenius, A.L., Toga,
Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W., Guan, Y., Shen, D., A.W., 2003. Mapping cortical change across the human life span. Nat. Neurosci.
Peng, Y., Zhang, D., Jie, Z., Wu, W., Qin, Y., Xue, W., Li, J., Han, L., Lu, D., Wu, P., 6, 309–315.
Dai, Y., Sun, X., Li, Z., Tang, A., Zhong, S., Li, X., Chen, W., Xu, R., Wang, M., Feng, Spear, L.P., 2000. The adolescent brain and age-related behavioral manifestations.
Q., Gong, M., Yu, J., Zhang, Y., Zhang, M., Hansen, T., Sanchez, G., Raes, J., Falony, Neurosci. Biobehav. Rev. 24, 417–463.
G., Okuda, S., Almeida, M., LeChatelier, E., Renault, P., Pons, N., Batto, J.M., Spear, L.P., 2011. Rewards, aversions and affect in adolescence: emerging
Zhang, Z., Chen, H., Yang, R., Zheng, W., Li, S., Yang, H., Wang, J., Ehrlich, S.D., convergences across laboratory animal and human data. Dev. Cognit. Neurosci.
Nielsen, R., Pedersen, O., Kristiansen, K., Wang, J., 2012. A metagenome-wide 1, 392–400.
association study of gut microbiota in type 2 diabetes. Nature 490, 55–60. Spear, L.P., 2013. Adolescent neurodevelopment. J. Adolesc. Health 52, S7–S13.
Rakic, P., Bourgeois, J.P., Goldman-Rakic, P.S., 1994. Synaptic development of the Stark, P.L., Lee, A., 1982. The microbial ecology of the large bowel of breast-fed and
cerebral cortex: implications for learning, memory, and mental illness. Prog. formula-fed infants during the first year of life. J. Med. Microbiol. 15, 189–203.
Brain Res. 102, 227–243. Steenbergen, L., Sellaro, R., van Hemert, S., Bosch, J.A., Colzato, L.S., 2015. A
Reiss, A.L., Abrams, M.T., Singer, H.S., Ross, J.L., Denckla, M.B., 1996. Brain randomized controlled trial to test the effect of multispecies probiotics on
development, gender and IQ in children. A volumetric imaging study. Brain cognitive reactivity to sad mood. Brain Behav. Immun. 48, 258–264.
119 (Pt 5), 1763–1774. Steinberg, L., Graham, S., O’Brien, L., Woolard, J., Cauffman, E., Banich, M., 2009. Age
Rhee, S.H., Pothoulakis, C., Mayer, E.A., 2009. Principles and clinical implications of differences in future orientation and delay discounting. Child Dev. 80, 28–44.
the brain-gut-enteric microbiota axis. Nat. Rev. Gastroenterol. Hepatol. 6, Steinberg, L., 2008. A social neuroscience perspective on adolescent risk-taking.
306–314. Dev. Rev. 28, 78–106.
Ringel-Kulka, T., Cheng, J., Ringel, Y., Salojarvi, J., Carroll, I., Palva, A., de Vos, W.M., Steinberg, L., 2010. A dual systems model of adolescent risk-taking. Dev.
Satokari, R., 2013. Intestinal microbiota in healthy U.S. young children and Psychobiol. 52, 216–224.
adults?a high throughput microarray analysis. PLoS One 8, e64315. Sturman, D.A., Moghaddam, B., 2011. The neurobiology of adolescence: changes in
Roberfroid, M.B., 2007. Prebiotics: the concept revisited. J. Nutr. 137 (Suppl. 2), brain architecture, functional dynamics, and behavioral tendencies. Neurosci.
830S–837S (3). Biobehav. Rev. 35, 1704–1712.
Roger, L.C., Costabile, A., Holland, D.T., Hoyles, L., McCartney, A.L., 2010. Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.N., Kubo, C., Koga, Y., 2004.
Examination of faecal Bifidobacterium populations in breast- and formula-fed Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal
infants during the first 18 months of life. Microbiology 156, 3329–3341. system for stress response in mice. J. Physiol. 558, 263–275.
Romeo, R.D., McEwen, B.S., 2006. Stress and the adolescent brain. Ann. N. Y. Acad. Tamminga, C.A., Buchsbaum, M.S., 2004. Frontal cortex function. Am. J. Psychiatry
Sci. 1094, 202–214. 161, 2178.
Romeo, R.D., Lee, S.J., Chhua, N., McPherson, C.R., McEwen, B.S., 2004. Testosterone Tana, C., Umesaki, Y., Imaoka, A., Handa, T., Kanazawa, M., Fukudo, S., 2010. Altered
cannot activate an adult-like stress response in prepubertal male rats. profiles of intestinal microbiota and organic acids may be the origin of
Neuroendocrinology 79, 125–132. symptoms in irritable bowel syndrome. Neurogastroenterol. Motil. 22,
Romeo, R.D., Bellani, R., Karatsoreos, I.N., Chhua, N., Vernov, M., Conrad, C.D., 512–519 (e114–e515).
McEwen, B.S., 2006. Stress history and pubertal development interact to shape Tarazi, F.I., Baldessarini, R.J., 2000. Comparative postnatal development of
hypothalamic-pituitary-adrenal axis plasticity. Endocrinology 147, 1664–1674. dopamine D(1), D(2) and D(4) receptors in rat forebrain. Int. J. Dev. Neurosci.
Romeo, R., Patel, R., Pham, L., So, V.M., 2016. Adolescence and the ontogeny of the 18, 29–37.
hormonal stress response in male and female rats and mice. In Special Issue on Tau, G.Z., Peterson, B.S., 2010. Normal development of brain circuits.
the Adolescent Brain. Neurosci. Biobehav. Rev. 70, 206–216. Neuropsychopharmacology 35, 147–168.
Romeo, R.D., 2003. Puberty: a period of both organizational and activational effects Tremaroli, V., Backhed, F., 2012. Functional interactions between the gut
of steroid hormones on neurobehavioural development. J. Neuroendocrinol. microbiota and host metabolism. Nature 489, 242–249.
15, 1185–1192. Tuohy, K.M., Fava, F., Viola, R., 2014. The way to a man’s heart is through his gut
Romeo, R.D., 2010. Adolescence: a central event in shaping stress reactivity. Dev. microbiota’ -dietary pro- and prebiotics for the management of cardiovascular
Psychobiol. 52, 244–253. risk. Proc. Nutr. Soc. 73, 172–185.
Roopchand, D.E., Carmody, R.N., Kuhn, P., Moskal, K., Rojas-Silva, P., Turnbaugh, P.J., Turnbaugh, P.J., Hamady, M., Yatsunenko, T., Cantarel, B.L., Duncan, A., Ley, R.E.,
Raskin, I., 2015. Dietary polyphenols promote growth of the gut bacterium Sogin, M.L., Jones, W.J., Roe, B.A., Affourtit, J.P., Egholm, M., Henrissat, B., Heath,
akkermansia muciniphila and attenuate high-fat diet-I nduced metabolic A.C., Knight, R., Gordon, J.I., 2009. A core gut microbiome in obese and lean
syndrome. Diabetes 64, 2847–2858. twins. Nature 457, 480–484.
312 K.-A. McVey Neufeld et al. / Neuroscience and Biobehavioral Reviews 70 (2016) 300–312

Tzounis, X., Rodriguez-Mateos, A., Vulevic, J., Gibson, G.R., Kwik-Uribe, C., Spencer, probiotic Bifidobacterium infantis 35624 in women with irritable bowel
J.P., 2011. Prebiotic evaluation of cocoa-derived flavanols in healthy humans syndrome. Am. J. Gastroenterol. 101, 1581–1590.
by using a randomized, controlled, double-blind, crossover intervention study. Williams, E.A., Stimpson, J., Wang, D., Plummer, S., Garaiova, I., Barker, M.E., Corfe,
Am. J. Clin. Nutr. 93, 62–72. B.M., 2009. Clinical trial: a multistrain probiotic preparation significantly
Van Leijenhorst, L., Gunther Moor, B., Op de Macks, Z.A., Rombouts, S.A., reduces symptoms of irritable bowel syndrome in a double-blind
Westenberg, P.M., Crone, E.A., 2010. Adolescent risky decision-making: placebo-controlled study. Aliment. Pharmacol. Ther. 29, 97–103.
neurocognitive development of reward and control regions. Neuroimage 51, Woo, T.U., Pucak, M.L., Kye, C.H., Matus, C.V., Lewis, D.A., 1997. Peripubertal
345–355. refinement of the intrinsic and associational circuitry in monkey prefrontal
Vazquez, D.M., Akil, H., 1993. Pituitary-adrenal response to ether vapor in the cortex. Neuroscience 80, 1149–1158.
weanling animal: characterization of the inhibitory effect of glucocorticoids on Wu, J.C., 2012. Psychological Co-morbidity in functional gastrointestinal disorders:
adrenocorticotropin secretion. Pediatr. Res. 34, 646–653. epidemiology, mechanisms and management. J. Neurogastroenterol. Motil. 18,
Vidal, J., Bie, J., Granneman, R.A., Wallinga, A.E., Koolhaas, J.M., Buwalda, B., 2007. 13–18.
Social stress during adolescence in Wistar rats induces social anxiety in Yakovlev, P.I., Lecours, A.-R., 1967. The myelogenetic cycles of regional maturation
adulthood without affecting brain monoaminergic content and activity. in the brain. In: Minkowski, A. (Ed.), Regional Development of the Brain in
Physiol. Behav. 92, 824–830. Early Life. Blackwell Scientific, Oxford, UK, pp. 3–70.
Vrieze, A., Van Nood, E., Holleman, F., Salojarvi, J., Kootte, R.S., Bartelsman, J.F., Yu, H.N., Zhu, J., Pan, W.S., Shen, S.R., Shan, W.G., Das, U.N., 2014. Effects of fish oil
Dallinga-Thie, G.M., Ackermans, M.T., Serlie, M.J., Oozeer, R., Derrien, M., with a high content of n-3 polyunsaturated fatty acids on mouse gut
Druesne, A., Van Hylckama Vlieg, J.E., Bloks, V.W., Groen, A.K., Heilig, H.G., microbiota. Arch. Med. Res. 45, 195–202.
Zoetendal, E.G., Stroes, E.S., de Vos, W.M., Hoekstra, J.B., Nieuwdorp, M., 2012. Yurkovetskiy, L., Burrows, M., Khan, A.A., Graham, L., Volchkov, P., Becker, L.,
Transfer of intestinal microbiota from lean donors increases insulin sensitivity Antonopoulos, D., Umesaki, Y., Chervonsky, A.V., 2013. Gender bias in
in individuals with metabolic syndrome. Gastroenterology 143, 913–916 autoimmunity is influenced by microbiota. Immunity 39, 400–412.
(e917).
Whorwell, P.J., Altringer, L., Morel, J., Bond, Y., Charbonneau, D., O’Mahony, L.,
Kiely, B., Shanahan, F., Quigley, E.M., 2006. Efficacy of an encapsulated

You might also like