You are on page 1of 17

Journal of Neuroendocrinology, 2014, 26, 707–723

© 2014 British Society for Neuroendocrinology


REVIEW ARTICLE

The Consequences of Early-Life Adversity: Neurobiological, Behavioural


and Epigenetic Adaptations
S. Maccari†, H. J. Krugers§, S. Morley-Fletcher†, M. Szyf¶ and P. J. Brunton**
†LIA, International Laboratory Associated, UMR 8576 CNRS Neural plasticity Team, University of Lille 1, France and Sapienza University of Rome, IRCCS
NEUROMED, Italy.
§Swammerdam Institute for Life Sciences, SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands.
¶Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.
**Division of Neurobiology, The Roslin Institute and R(D)SVS, University of Edinburgh, Midlothian, UK.

Journal of During the perinatal period, the brain is particularly sensitive to remodelling by environmental
Neuroendocrinology factors. Adverse early-life experiences, such as stress exposure or suboptimal maternal care, can
have long-lasting detrimental consequences for an individual. This phenomenon is often referred
to as ‘early-life programming’ and is associated with an increased risk of disease. Typically,
rodents exposed to prenatal stress or postnatal maternal deprivation display enhanced neuroen-
docrine responses to stress, increased levels of anxiety and depressive-like behaviours, and cog-
nitive impairments. Some of the phenotypes observed in these models of early-life adversity are
likely to share common neurobiological mechanisms. For example, there is evidence for impaired
glucocorticoid negative-feedback control of the hypothalamic-pituitary-adrenal axis, altered glu-
tamate neurotransmission and reduced hippocampal neurogenesis in both prenatally stressed
rats and rats that experienced deficient maternal care. The possible mechanisms through which
maternal stress during pregnancy may be transmitted to the offspring are reviewed, with special
consideration given to altered maternal behaviour postpartum. We also discuss what is known
about the neurobiological and epigenetic mechanisms that underpin early-life programming of
Correspondence to: the neonatal brain in the first generation and subsequent generations, with a view to abrogating
P. J. Brunton, Division of programming effects and potentially identifying new therapeutic targets for the treatment of
Neurobiology, The Roslin Institute and stress-related disorders and cognitive impairment.
R(D)SVS, University of Edinburgh,
Easter Bush, Midlothian EH25 9RG, Key words: cognition, DNA methylation, hippocampus, HPA axis, maternal behaviour, mood dis-
UK orders, prenatal stress
(e-mail: p.j.brunton@ed.ac.uk).
doi: 10.1111/jne.12175

this programming is maladaptive, increasing the susceptibility of


Introduction
an individual to adulthood pathologies such as cardiovascular
It is widely accepted that the physiological and behavioural traits disease, metabolic syndrome, cognitive impairments and affective
of an individual are determined by complex interactions between disorders (6). The idea that adult vulnerability to disease may be
their genes (‘nature’) and their experiences/environment (‘nurture’). programmed or imprinted during the foetal period (i.e. foetal
The perinatal period, infancy, childhood and puberty are periods programming) was first proposed by Barker et al. (7,8) and is
of increased neuroplasticity and therefore the brain is particularly now widely accepted. This idea has subsequently been extended
sensitive to remodelling by environmental factors at these times. to also include the early postnatal period, termed ‘perinatal pro-
Exposure to adversity (such as stress or maternal deprivation) in gramming’, where nongenetic factors act in early life (pre- or
early-life can ‘programme’ or ‘imprint’ persistent neuroendocrine, postnatally) to permanently organise or imprint physiological sys-
behavioural and metabolic changes (1–4). These programming tems.
effects may be considered adaptive, permitting an organism to Early adverse experiences can programme adult health in two
adapt to cope with unfavourable conditions (5). However, often ways: either by cumulative damage over time or by the biological
708 S. Maccari et al.

embedding of adversities during sensitive developmental periods. required and the intensity of the stress is more difficult to maintain
For example, according to the developmental hypothesis of mood at a consistent level between animals and across time, in contrast
disorders, stressful events occurring during critical periods of brain to the PRS model.
development trigger a maladaptive programme that alters mecha-
nisms of resilience to stress across the entire lifespan. Along this
Prenatal stress and hypothalamic-pituitary-adrenal (HPA)
line, major depression can be seen as a latent outcome of stressful
axis dysfunction in the offspring
early-life events (9–15).
This review will focus on adversity in the perinatal period. We A key feature of both of these models is a permanent impairment
first discuss the consequences of prenatal stress on the offspring’s of HPA axis functioning in the offspring (Table 1). PRS offspring
brain and behaviour (taking into account sex differences), the possi- exhibit prolonged corticosterone responses to stress in both male
ble central mechanisms underpinning these altered phenotypes and and female PRS rats (4,26,28). Similarly, both male and female PSS
how prenatal stress effects may be transmitted to the developing offspring display enhanced and prolonged HPA axis responses to
foetus. Next, we consider the importance of the early postnatal physical (Fig. 2) and psychological stressors in adulthood (1).
environment, in particular the quality of maternal care on brain Prenatal restraint stress (PRS) also affects age-related alterations
development of the neonate. Finally, we review the evidence for in HPA axis function. For example, the HPA axis period of hypore-
early-life adversity in transgenerational programming of future gen- sponsiveness typically observed in the early postnatal period (29)
erations and discuss the epigenetic mechanisms involved. is abolished in newborn PRS rats (16) and circulating glucocorti-
coid levels in middle-aged PRS animals are similar to those found
in aged nonstressed animals (30). Moreover, PRS alters not only
Consequences of prenatal stress on the brain and
reactive adaptation, but also predictive adaptation by altering cir-
behaviour in later life
cadian rhythmicity. Both male and female PRS rats have greater
The use of early-life environmental manipulations in animal models levels of corticosterone at the end of the light period, and females
has spawned the study of individual vulnerability to stress-related display elevated corticosterone levels over the entire diurnal cycle
disorders, which should aid the development of new therapies and (31). The sleep–wake cycle is also dramatically modified by PRS
enhance our understanding in such a way as to improve prevention (32,33), with a significant increase in the amount of rapid eye
strategies. A plethora of studies have investigated the consequences movement sleep over a 24-h recording session, which is positively
of manipulating the prenatal environment (e.g. by maternal stress correlated with plasma corticosterone concentrations. Significant
exposure, maternal glucocorticoid administration, maternal nutrient phase advances are also observed in the circadian rhythms of
restriction) on the developing offspring. The majority of these stud- locomotor activity relative to the entrained light/dark cycle in both
ies have been performed in rodents and multiple paradigms of male and female PRS rats. Furthermore, when subjected to an
‘adversity’ have been reported in the literature. Pregnant rats, mice abrupt shift in the light/dark cycle, PRS rats resynchronise their
or guinea pigs are typically repeatedly exposed to a either a psy- activity rhythm to a new light/dark cycle slower than control rats
chological stressor (e.g. repeated restraint, noise or strobe lighting) (34).
(16–18), a physical stressor (e.g. hypoxia or immune challenge)
(19,20) or a combined psychological and physical stressor (e.g.
Prenatal stress and affective behaviour in the offspring
social stress) (1,21) either throughout or during a specific period of
gestation. Alternatively, chronic variable stress paradigms are also The hyperactivity of the HPA axis observed in PRS and PSS off-
commonly utilised that involve exposing pregnant animals to a spring is accompanied by increased anxiety- and depressive-like
combination of different stressors on an unpredictable basis behaviour during the life span (Table 1). This is evidenced in male
(22–24). PRS offspring (females were not studied) by increased ultrasonic
Here, we focus on two prenatal stress paradigms in the rat, with vocalisations (USVs) in infancy (35), reduced social play during ado-
which we have most experience: the prenatal restraint stress (PRS) lescence (36), reduced exploration in the elevated-plus maze test
model and the prenatal social stress (PSS) model. Both are well- and open field test (30,37) and increased immobility in the forced-
documented models of early-life stress (ELS) (4,25). The PRS model swim test during adulthood (27,38–40). Rats subjected to PRS emit
(26) involves restraining a pregnant rat in a transparent Plexiglas significantly more USVs in response to isolation at postnatal day 10
cylinder, three times a day for 45 min under bright light: from day compared to controls. In addition, PRS pups do not show the phe-
11 of pregnancy until term (4,27) (Fig. 1), whereas the PSS model nomenon of ‘maternal potentiation’ (i.e. the increase in USVs in
involves transferring a pregnant rat to the cage of an unfamiliar response to a brief maternal reunion after isolation), normally seen
(and hence aggressive) lactating rat for 10 min/day for 5 consecu- at postnatal day 10 and postnatal day 14 (35).
tive days (gestation days 16–20) during the last week of pregnancy Adult male PSS offspring also display increased anxiety-related
(1). The advantage of the PSS model is that it is considered etho- behaviour on the elevated plus maze (1). However, PSS does not
logically relevant and, because of the ‘social’ aspect, it perhaps bet- appear to affect anxiety-type behaviour in the female offspring
ter reflects the types of stress pregnant women are likely to (1). A distinct sex difference in anxiety-like behaviour is also
experience. However, because this model relies on the behaviour observed in offspring of the PRS model, with males appearing to
exhibited by lactating rats (e.g. aggression), more animals are be more susceptible than females (41). Taken together, these data

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 709

Reduced
Prenatal
maternal
restraint stress
behaviour Anxiety and glutamate
35
CON r = 0.9
PRS P < 0.01
30

Time spent in open arms (%)


25

20

15

10

0
100 200 300 400
Depolarisation-stimulated glutamate release
(pmol/mg prot.)

E11 P0 P21 3 months


Pregnancy Birth Weaning Adulthood

Fig. 1. Prenatal restraint stress (PRS) in the rat: a model of early programming of stress-related diseases from molecules to behaviour. During the second half
of gestation, the dam is restrained (45 min 9 3 times/day) under bright light. The procedure is not painful but the dam cannot escape. The offspring and the
mother are then left undisturbed until weaning (P21). Gestational stress reduces subsequent maternal behaviour and the adult PRS offspring are characterised
by enhanced anxiety and reduced glutamate release in the ventral hippocampus. Based on data from Marrocco et al (54). CON, control; E, embryonic day; P,
postnatal day; PRS, prenatal restraint stress.

offer a clear demonstration that prenatal stress exposure causes vulnerable than females, and this effect is observed mainly in the
anxiety and depression-like behaviour in the male offspring, which ventral hippocampus (41). However, in addition to a sex-dependent
is already evident early in development, but also depression-like effect, alterations in neuroplasticity may also depend on the genetic
behaviour in female PRS rats (40). Chronic treatment with anti- background of the individual. Indeed, prenatal stress has been
depressants in adulthood can fully reverse the key alterations shown to differentially alter the consequences of a genetic predis-
induced by PRS (33,38,39), indicating that models of prenatal position to either high- or low-anxiety-related behaviour on neuro-
stress, such as the PRS paradigm, can replicate the developmental plasticity. Prenatal stress results in reduced neurogenesis in rats
factors involved in the etiology of anxious/depressive disorders with genetically determined high emotionality but not those with
(14), and have predictive and face validity as an experimental ani- low emotionality (48,49). The PRS-induced deficit in neurogenesis
mal model of anxiety and depression in humans (10). can be restored by chronic treatment with antidepressants such as
fluoxetine and agomelatine (39) or the neurotrophin, insulin-like
growth factor-1 (45).
Prenatal stress, impaired cognition and related
neuroplasticity in the offspring
Central mechanisms underlying altered phenotypes in
Studies in nonhuman primates and rodents have reported altera-
prenatally stressed offspring
tions in the structure and function of the hippocampus as a conse-
quence of prenatal stress (42,43) and several studies have shown Because the brain is particularly sensitive to programming by pre-
learning and/or memory impairment in the offspring of dams natal stress exposure and programming of the brain is associated
stressed during pregnancy (41,44). However, the timing of the pre- with several negative phenotypes (e.g. anxiety, depression, impaired
natal stress (early versus mid- or late-gestation) (44), the type and/ cognition), understanding the central mechanisms involved is
or the severity of the prenatal stress, the sex of the offspring (41) important to pave the way for new therapeutic strategies and iden-
and the age of the offspring at time of memory assessment (e.g. tify potential pharmacological targets.
juvenile, adult or aged) appear to be critical. PRS during late gesta-
tion generally has little effect on cognitive performance in the
Increased forward drive and impaired negative-feedback
Morris water maze and Y-maze in young male adults (30,41)
control of the HPA axis
(Table 1) but exacerbates memory impairment observed during age-
ing in these tests (30,45,46). Cognitive deficits induced by PRS are Enhanced adrenocorticotrophic (ACTH) and corticosterone responses
linked to a reduction in hippocampal neurogenesis (42,47). In to stress in PSS offspring are associated with elevated expression
particular, these effects are sex-dependent, with males being more of mRNA for the ACTH secretagogues, corticotrophin-releasing

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
710 S. Maccari et al.

Table 1. Comparison of Neuroendocrine and Behavioural Effects of the Pre-


natal Restraint Stress (PRS) and Prenatal Social Stress (PSS) Models. Control PSS

PRS PSS

Parameter Male Female Male Female References


pPVN CRH
Basal corticosterone ↑* ↑ ↔ ↑ 1,28 mRNA
levels
Stress-induced ↑ ↑ ↑ ↑ 1,16,26
corticosterone levels
Hippocampal GR ↓† NA ↔‡ ↔‡ 1,16,26 300
Hippocampal MR ↓† NA ↓‡ ↓‡ 1,16,26

Δ ACTH 30 min post-IL-1β (pg/ml)


Anxiety-type ↑ ↓ ↑ ↔ 1,30,35–37,41 *
250
behaviour
Depressive-like ↑ ↑ NA NA 27,38–40 200
behaviour
Spatial memory ↔ NA ↔ ↔ 30,37, Brunton PJ Anterior 150 ACTH
(young rats) and Lai Y-T, pituitary
unpublished 100
data
Working memory ↔ NA NA NA 30,37 50
(young rats)
Working memory ↓ NA NA NA 30 0
(aged rats) CON PSS

Δ Corticosterone 60 min post-IL-1β (ng/ml)


250
The neuroendocrine and behavioural effects of the PRS and PSS models on *
the male and female offspring compared to age-matched control offspring
(mothers undisturbed during pregnancy). ↑, increase; ↓, decrease or ↔, no 200
significant effect. GR, glucocorticoid receptor, MR, mineralocorticoid recep-
tor; *Only at end of light phase; †receptor binding; ‡mRNA expression. NA, 150
Adrenal
no data available. CORT
cortex
hormone (CRH) and vasopressin in the paraventricular nucleus 100
(PVN), indicating increased excitatory drive to the anterior pituitary
by the hypothalamus (Fig. 2). Whether this is a result of enhanced 50
drive to the PVN CRH neurones, reduced inhibitory input to the
CRH neurones or a combination of both is not clear. 0
CON PSS
The HPA axis is under negative-feedback control by glucocortic-
oids, acting via glucocorticoid (GR) and mineralocorticoid (MR)
Fig. 2. Enhanced stress-induced hypothalamic-pituitary-adrenal (HPA) axis
receptors. Prolonged corticosterone responses to stress in PRS
responses in prenatally stressed rats. HPA axis responses in adult male off-
offspring occur concomitant with reduced levels of both MR and spring born to control (CON) dams or dams exposed to social stress (PSS)
GR in the hippocampus, which is evident in the neonate, adoles- from days 16–20 of gestation. Rats were blood sampled before and after
cent and adult (16,26). Similarly, expression of MR mRNA is treatment with the cytokine, interleukin-1b (IL-1b; 0.5 lg/kg i.v.) and then
reduced in the hippocampus in adult male and female PSS off- killed 4 h later. Neurones located in the parvocellular region of the paraven-
spring (1). Together, these findings indicate impaired glucocorti- tricular nucleus (pPVN) in the hypothalamus express corticotrophin-releasing
hormone (CRH) mRNA (detected by in situ hybridisation). IL-1b-induced
coid negative-feedback control of the HPA axis in prenatally
CRH mRNA levels are significantly greater in the PSS rats compared with
stressed offspring.
controls, resulting in a three-fold greater secretion of adrenocorticotrophic
The mechanisms through which hippocampal GR and/or MR may hormone (ACTH) from the anterior pituitary and corticosterone (CORT) secre-
be down-regulated are not fully understood, although they likely tion from the adrenal cortex. *P < 0.001, Student’s t-test. Based on data
involve epigenetic changes, as has been previously demonstrated from Brunton and Russell (1).
for altered GR gene expression in the hippocampus of rats exposed
to varying degrees of postnatal maternal care (discussed below)
(50) and in mice exposed to prenatal stress (23). may be effective in correcting the pathological phenotype caused
by ELS. Alterations in glutamate neurotransmission are assumed to
play a role in the pathophysiology of stress-related disorders
Glutamate neurotransmission in the hippocampus
(51–53). Indeed, a long-term alteration in glutamate transmission
Therapeutic strategies aimed at restoring the balance between lies at the core of the neuroplastic programming and anxious
excitatory and inhibitory neurotransmission in the hippocampus phenotype induced by PRS. PRS induces a selective reduction in

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 711

glutamate release in the ventral hippocampus that is causally


Transmission of prenatal stress effects from mother to
related with the enhanced anxiety-like behaviour displayed by PRS
foetus
animals (Fig. 1). Furthermore, pharmacological enhancement of
glutamate release in the ventral hippocampus abolishes this
Glucocorticoids
behavioural pattern (54). PRS rats also show reduced expression
and function of group-I and group-II hippocampal metabotropic The mechanisms responsible for transmitting the effects of maternal
glutamate (mGlu) receptors, with a marked reduction of mGlu1 stress to the foetus(es) remain unclear; however, given that an
and mGlu5 observed selectively in males and a reduction in mGlu important feature of the stress response is the secretion of glucocor-
2/3 in both males and females (35,41,46). The alterations in mGlu ticoids, these steroids have become the preferred candidate
receptors in the hippocampus induced by PRS can be detected at ‘programming factor’ conveying maternal stress effects to the foetus
infancy with mGlu5 and mGlu1 receptors reduced in PRS rats by (es) (66,67). The evidence is as follows: (i) pregnant rodents treated
postnatal day 10, whereas expression of mGlu2/3 receptors with the synthetic glucocorticoid, dexamethasone, produce offspring
declines only after weaning (35). with phenotypes similar to prenatally stressed offspring (18,68) and
Thus, glutamatergic transmission in the ventral hippocampus, a (ii) maternal adrenalectomy (to remove the source of maternal gluco-
key brain region involved in the (mal)adaptive programming caused corticoids) abrogates some of the effects of PRS in the offspring (69).
by ELS, represents an attractive pharmacological target for novel Nevertheless, a role for maternal glucocorticoids in transmitting
therapeutic strategies in the treatment of depression and anxiety the effects of maternal stress to the foetus is not clear-cut. First, the
(10,54,55). Indeed, chronic treatment with fluoxetine or agomelatine placenta expresses 11b-hydroxysteroid dehydrogenase type-2
can reverse the depressive-like phenotype induced by PRS by (11b-HSD2), which serves to limit foetal exposure to maternal gluco-
enhancing glutamatergic transmission in the ventral hippocampus corticoids by metabolising corticosterone into inactive 11-dehydro-
(56). corticosterone (70). However, repeated stress exposure in pregnancy
significantly reduces activity of this barrier enzyme and placental
gene expression for 11b-HSD2 (71,72), which means potentially
Anxiety and altered CRH receptor expression in the
increased exposure of the foetuses to maternal glucocorticoids. Sec-
amygdala
ond, maternal HPA axis responses to stress are dramatically reduced
The amygdala CRH system plays a key role in mediating anxiety- in late pregnancy, which is assumed to minimise foetal exposure to
like behaviours (57–59). Increased levels of CRH mRNA are maternal glucocorticoids (73). Indeed, PSS offspring display pro-
observed in the central nucleus of the amygdala (CeA) of both grammed phenotypes despite the maximal maternal corticosterone
male and female PSS offspring (1). This increase in CRH is probably response during the PSS exposure not exceeding the concentrations
facilitated by glucocorticoids because glucocorticoids up-regulate observed at the diurnal peak in gestation (74). However, it may be
CRH gene expression in the amygdala (60) and central GR facilitate that repeated stress, in combination with reduced effectiveness of
anxiety-type behaviours. Indeed, GR mRNA expression is also up- placental 11b-HSD2, leads to exposure of the foetus(es) to maternal
regulated in the CeA in PSS rats (1). However, this is unlikely to glucocorticoids at levels sufficient to programme the offspring.
explain the anxiety-like behaviour in PSS male rats because both Finally, it should be noted that, in foetal rats, the adrenal glands
sexes show elevated CRH and GR mRNA expression in the CeA, are capable of corticosterone secretion from embryonic day 16.
whereas only the males exhibit an anxious phenotype (1). Never- Because maternal stress in late pregnancy results in activation of
theless, there are sex differences in CRH receptor expression in the the foetal HPA axis (75,76), it is plausible that glucocorticoids of
amygdala in PSS offspring indicating that these changes may be foetal origin may also contribute to programming the foetal brain.
critical in the expression of an anxious phenotype (61). Two dis-
tinct receptors mediate the actions of CRH: the type 1 receptor
Catecholamines
(CRH-R1) mediates anxiogenic behavioural responses, whereas the
type 2 receptor (CRH-R2) mediates the anxiolytic actions of Maternal adrenalectomy not only removes the source of glucocor-
urocortins 2 and 3 (62). In PSS males, CRH-R1 is increased in the ticoids, but also removes the source of other hormones (e.g. cate-
CeA and basolateral amygdala, whereas CRH-R2 is decreased in cholamines, sex steroids, mineralocorticoids), which could
the basomedial amygdala; however this effect is not seen in PSS potentially contribute to foetal programming. By contrast to stress-
females (61). This altered ratio of CRH-R1 to CRH-R2 expression in induced adrenaline secretion from the adrenal medulla, which is
the amygdala in PSS males may contribute to increased anxiogenic attenuated in late pregnancy, noradrenaline responses to stress are
actions of CRH, and decreased anxiolytic actions of urocortins 2 maintained in pregnancy (77,78). Elevated levels of circulating
and/or 3, thus explaining the anxious-like behaviour observed in maternal catecholamines have the potential to negatively influence
PSS males. Indeed, heightened anxiety has been associated with foetal development: vasoconstriction of placental blood vessels will
increased CRH-R1 mRNA expression in the PVN (63) and reduced reduce placental blood supply, thus impairing the delivery of oxy-
CRH-R2 expression in the amygdala (64) in other models of prena- gen and nutrients to the foetuses. Given that both hypoxia and
tal stress in rats, and forebrain CRH-R1 signalling is necessary for nutrient restriction activate the foetal HPA axis (79,80) and the foe-
the expression of anxiogenic behaviour in a mouse model of ELS tal sympathetic-adrenomedullary system (81), these responses may
(65). also contribute to programming of the offspring (19,63,82,83).

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
712 S. Maccari et al.

adoption modifies maternal behaviour, increasing pup-directed


Altered placental function
behaviour in foster mothers, and decreasing stress-induced cortico-
Foetal growth and development relies on adequate placental nutri- sterone secretion in adult PRS offspring (26). Moreover, stress-
ent transfer; thus, placental dysfunction could also be implicated in induced impairments in subsequent maternal behaviour (following
programming the offspring. A key nutrient for the developing foe- repeated restraint stress during the second half of pregnancy) can
tus is glucose, which crosses the placental barrier via transporters be reversed by treating the lactating dam with oxytocin receptor
(84). Glucose transporter type 1 (GLUT1) is the most abundantly agonists (Mairesse J, Gatta E, Reynaert ML, Marrocco J, Morley-
expressed of the glucose transporters in rodent placentas (85) and Fletcher S, Soichot M, Deruyter L, Van Camp G, Bouwalerh H, Fagioli
is considered rate limiting for glucose transport from mother to F, Pittaluga A, Allorge D, Nicoletti F, Maccari S; unpublished data),
foetus. Placental expression of GLUT1 is decreased by PRS and is highlighting the possibility of targeting the central oxytocinergic
associated with reduced levels of glucose and growth hormone in system in the treatment of stress-related disorders both in the
the foetal circulation (72). As noted above, placental expression and adult offspring and in the mothers during the postpartum period
activity of 11b-HSD2 is also strongly reduced by PRS (72), which (93, Mairesse J, Gatta E, Reynaert ML, Marrocco J, Morley-Fletcher
would be expected to lead to increased transfer of maternal gluco- S, Soichot M, Deruyter L, Van Camp G, Bouwalerh H, Fagioli F, Pit-
corticoids to the foetal compartment (70). taluga A, Allorge D, Nicoletti F, Maccari S; unpublished data). There
Interestingly, placental expression of 11b-HSD2 is selectively are, however, some important considerations that should be taken
increased in rats with genetically determined low anxiety-related into account when manipulating the oxytocin system, particularly
behaviour (49) but not in rats with high anxiety-related behaviour, in the peripartum period (94).
thus indicating that the gene 9 early environment interaction con-
tributes to shape placental function. Together, these changes in pla-
Consequences of postnatal stress on the brain and
cental function induced by ELS may underlie reduced birth weight
behaviour in later life
observed in PRS and PSS offspring and contribute to programming
of metabolism, inflammatory responses, the brain and behaviour In rodents, the presence of the mother during the initial weeks
(1,2,72,86). after birth is of fundamental importance for the offspring. During
Epigenetic mechanisms are critically involved in determining pla- this time, the interaction between mother and pups is highly rele-
cental functioning and a recent study by Howerton et al. (86) has vant for nutrition, body temperature and physiological homeostasis
identified O-linked-N-acetylglucosamine (O-GlcNAc) transferase (95). Postnatally, the brain is still developing and disturbances of
(OGT), as a promising placental biomarker of maternal stress expo- the early postnatal mother–pup relationship can have major and
sure (early gestation PRS in mice). OGT is an X-linked gene impor- long-lasting consequences for neuroendocrinology, neuronal func-
tant in regulating proteins involved in chromatin remodelling. tion, behaviour and adaptation of the offspring. Moreover, there is
Levels of both OGT and its biochemical marker, O-GlcNAcylation, increasing evidence from studies in biparental species that paternal
are significantly lower in males than females and are further deprivation can have similar adverse neurobiological and behaviour-
reduced by prenatal stress exposure (86). Moreover, examination of al consequences for the offspring (96).
human placental tissue has found similar patterns related to X
chromosome dosage (86). Hypothalamic gene expression and the
Postnatal stress and HPA axis dysfunction in the offspring
broad epigenetic microRNA environment in the neonatal brain of
placental-specific hemizygous OGT mice are markedly altered, high- The presence of the dam during the early postnatal period is crucial
lighting the importance of placental OGT in neurodevelopment (86). for maintaining activity of the HPA axis (88,89). In general, the
Interestingly, OGT interacts with the GR to repress gene expression presence of the dam results in lower levels of HPA axis activity in
(87), thus strengthening the epigenetic role played by placental OGT the offspring (97–101). During the first 2 weeks of life, pups show
in the programming induced by prenatal stress. diminished reactivity of the HPA axis in response to mild–moderate
stressors (stress hyporesponsive period; SHRP) (29,102). Disruption
of the early-life environment and the SHRP by separating pups
Altered maternal behaviour
from the dam has substantial and long-lasting neuroendocrine and
The early postnatal environment is known to influence the activity behavioural consequences. In general, maternal deprivation and
of the HPA axis and anxiety-like behaviour in later life (88,89). maternal separation result in phenotypes similar to those observed
Given that stress exposure during gestation can alter subsequent in prenatally stressed offspring (described above), including HPA
maternal behaviour (90,91) and increase anxiety-related behaviour axis hyperactivity (103–108), as well as altered social behaviour and
postpartum (92), it is important to take this into account when increased aggression (109).
considering how the effects of maternal stress during pregnancy
are transmitted from the mother to the offspring. Although the
Postnatal stress, impaired cognition and related
importance of maternal care for offspring development is discussed
neuroplasticity in the offspring
in detail below, it is important to note that prenatal stress-related
outcomes on HPA axis function in the offspring can be reversed by Postnatal stress also results in cognitive impairments (110–112)
early postnatal manipulation such as adoption (26). Indeed, and higher levels of anxiety (109,113–115). Moreover, hippocampal

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 713

structure is affected: maternal separation and deprivation has been In parallel with increased dendritic complexity, LTP can easily be
found to decrease mossy fibre density (116), reduce neurogenesis induced in the CA1 area and dentate gyrus in high LG-ABN off-
(112,117–119), reduce brain-derived neurotrophic factor expression spring but not in low LG-ABN offspring (135,137,139). Recent stud-
(120,121) and alter the number of primary dendrites in dentate ies have also described that synaptic currents are altered by
gyrus granule cells in a sex-dependent manner (an increase is maternal care. NMDA currents are enhanced in the dentate gyrus
found in females and a decrease in males) (112,122). Similar of offspring of low LG-ABN animals and maternal deprivation
findings are observed in mice that were exposed to chronic ELS affects synaptic expression of NMDA receptors (140). Interestingly,
evoked by reducing nesting and bedding material between P2 and application of NMDA receptor antagonist enabled synaptic plasticity
P9 (123,124). ELS results in elevated plasma corticosterone levels in in offspring of low LG-ABN animals. Although paradoxical, these
adult animals as a result of enhanced excitatory glutamatergic studies may suggest that enhanced NMDA receptor function under-
transmission in the PVN (125), together with reduced spatial learn- lies the reduced ability to elicit hippocampal LTP in low LG-ABN
ing, impaired object recognition, reduced hippocampal dendritic offspring. In agreement with an impaired ability to express synaptic
arborisation and disrupted hippocampal long-term potentiation plasticity, low LG-ABN animals show spatial learning deficits in the
(LTP) (123,124,126). These cognitive deficits and structural changes Morris water maze (134) and impaired object recognition memory
in the hippocampus induced by ELS appear to be dependent, at (141) compared to high LG-ABN offspring.
least in part, on forebrain CRH-R1 signalling (126,127). Not only neuroendocrine regulation and synaptic plasticity are
affected by maternal care, but also the synaptic function of low
LG-ABN and high LG-ABN animals appears to be differently regu-
Maternal care, cognition and neuroplasticity in the
lated by stress hormones. Thus, synaptic plasticity in low LG-ABN
offspring
animals is facilitated by bath-application of corticosterone and/or
Maternal deprivation and maternal separation disturb mother– noradrenaline to the slices (135,137,139). This indicates that synapses
infant interactions in a complex way. During separation and depri- of low LG-ABN animals do have the ability to express synaptic plas-
vation, the offspring are deprived of maternal care (such as licking/ ticity and that the sensitivity of synapses is determined by variations
grooming, nutrition, warmth) for a shorter or longer period of time. in postnatal maternal care (Fig. 3). Accordingly, memory formation in
However, upon reunion with the mother, the pups may actually a highly emotional contextual fear conditioning task is markedly
receive enhanced levels of maternal care (112). Because maternal improved in low LG-ABN animals compared to high LG-ABN off-
separation and deprivation are accompanied by alterations in spring (135). Similar effects have been found after maternal depriva-
maternal behaviour (88,128–130), it is highly relevant to understand tion (112). These observations indicate that low levels of postnatal
how naturally occurring variations in maternal care provided by the maternal care renders synapses more efficient under stressful condi-
dam affects the offspring (131). tions and enhances memory formation for emotionally arousing
Studies have explored this behavioural variability in a normal, events, which may be an example of behavioural adaptation (142).
undisturbed population of Long Evans rats. It was confirmed that Interestingly, recent studies have demonstrated that there is con-
substantial differences in maternal care exist between dams (88) and siderable variation in maternal care even within a litter, which also
that this trait was stable over time and over multiple litters (132).
More specifically, licking and grooming (LG) and arched-back nursing 250 Vehicle
(ABN) of the pups by the dam were shown to predict later-life phe- Cort
Synaptic potentiation (% of baseline)

notypic outcome (88,133–135). The offspring of low LG-ABN dams


200
exhibit enhanced HPA axis responses to stress in adulthood, *
increased CRH mRNA levels in the hypothalamus and reduced hippo-
campal GR levels (88,135). Hippocampal GR expression is epigeneti- 150
cally programming via maternal care as discussed later. Importantly, *
these neuroendocrine effects of variations in maternal care are
reversed by cross-fostering, thereby directly implying the relevance 100
of maternal care in neuroendocrine regulation of the HPA axis (88).
Different levels of maternal care during the early postnatal per-
50
iod can also regulate hippocampal structure. Neuronal survival in
the adult dentate gyrus (136), dendritic length of hippocampal CA1
pyramidal cells and dentate gyrus granular cells, the spine number 0
of hippocampal CA1 pyramidal cells (135) and dentate gyrus granu- L-LG H-LG
lar cells (137) and synaptophysin and neural cell adhesion molecule
Fig. 3. Maternal care determines sensitivity to corticosterone (Cort). Synap-
levels (88) are all increased in adult offspring from high LG-ABN
tic potentiation is enhanced in high licking/grooming (H-LG) animals com-
dams. However, dendritic complexity and spine density in layer II/III pared to low licking/grooming (L-LG) animals. However, corticosterone
neurones of the somatosensory cortex were found to be reduced in enhances synaptic potentiation in L-LG animals at the same time as sup-
high LG-ABN offspring (138) indicating that maternal care can have pressing synaptic plasticity in H-LG animals. *P < 0.05, Student’s t-test.
different effects on dendritic complexity in different brain areas. Based on data from Champagne et al. (135).

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
714 S. Maccari et al.

has long-lasting consequences for brain and behaviour (143–145). Early-life programming of the genome: the role of
In the hippocampus, the pup LG-ABN score positively correlates epigenetics
with GR mRNA expression (145). In addition, the ability to evoke
There is a strong body of data supporting the hypothesis that ELS
synaptic plasticity in the dentate gyrus correlates positively with
results in long-term and stable phenotypic changes that involve per-
high LG-ABN scores although dendritic morphology in the dentate
sistent alterations in gene function. This begs the question of what
gyrus correlates less well with LG-ABN scores and differs between
comprises the mechanism that embeds the effects of early-life
males and females (145). Moreover, LG-ABN scores result in slight
adversity in the genome. Although much attention in understanding
effects on adult CA1 structure and function, which appear to be
how malfunctioning genes are involved in neuropathology in the last
sex-dependent (144).
decades has focussed on rare or common sequence alterations, the
Within-litter variations of maternal care also correlate with
data reviewed above indicate the possibility that gene function might
behaviour in adult animals. Thus, LG-ABN scores during early life
also be stably altered by the early environment and particularly by
correlates positively with behavioural expressions of social play
behavioural exposure. The classical idea of gene–environment inter-
(143) and decision making and correlates negatively with anxiety in
action focussed on how risk alleles affect the phenotype under dif-
rats (146).
ferent environments. However, these studies do not offer a
In conclusion, there is considerable evidence that maternal care
mechanism for this ‘magical’ interaction of environment and genes
during the early postnatal period can have long-lasting conse-
and the focus on polymorphic risk alleles might lead to the idea that
quences for brain structure, synaptic function, responsiveness of
‘gene–environment’ effects are limited to polymorphic genes (154).
synapses and behaviour.
Both the persistence and commonality of the effects of early-life
adversity suggest that physiological mechanisms must exist that
Transgenerational effects of early-life adversity translate these exposures to persistent gene function differences and
that they act on the common genetic landscape.
In recent years, research on the influence of early-life adversity has
The challenge to the idea that genotype exclusively defines phe-
been extended to include not only the first-generation offspring,
notypes was first addressed in the field of embryology when it
but also subsequent generations. It has become increasingly clear
became clear that, in multicellular organisms, such as mammals,
that the detrimental effects of early-life adversity (described above)
the same genetic heritage common to all different tissues expresses
can be transmitted to future generations, apparently via nonge-
numerous different phenotypes. Waddington (155) was the first to
nomic mechanisms (discussed below). Maternal under- or over-
coin the term ‘epigenetics’ to describe the persistent differentiation
nutrition during pregnancy has been most extensively studied. In
of gene function during embryonic development. The idea was that
guinea pigs, the effects of maternal under-nutrition during preg-
the genes undergo ‘nongenetic’ changes in their mode of function
nancy on HPA axis activity in the offspring are transmitted to both
that would define both the spatial and temporal profile of gene
the first- and second-generation offspring (147) and, in mice, phe-
function in the developing organism (155). Several decades of
notypes resulting from maternal high-fat diet (e.g. body length and
research have unravelled the epigenetic mechanisms that differenti-
insulin insensitivity) can be passed to subsequent generations
ate gene function during embryonic development. However, these
(148,149). Moreover, in rats, the effect of maternal administration
mechanisms respond to the innate signals of embryonic develop-
of dexamethasone during pregnancy on glucose tolerance is trans-
ment. It is easy to see how exposure to chemicals that intervene
mitted to the second-generation offspring via either the maternal
with epigenetic processes during embryonic development will dis-
(i.e. where prenatally dexamethasone-exposed F1 females are bred
rupt the generation of the epigenetic profile and will result in long-
with na€ıve males) or paternal line (i.e. where prenatally dexametha-
term changes in the expression of genes in the brain and other tis-
sone-exposed F1 males are bred with na€ıve females) (150). Recent
sues. The main question with response to early-life adversity is
studies have demonstrated that paternal stress exposure prior to
whether the same mechanism that differentiates gene function
conception can programme altered stress responsivity (151) and
during embryology in response to ‘innate’ signals will also respond
behaviour (152,153) in the subsequently conceived adult offspring,
to ‘external’ signals from the environment and whether behavioural
and this is associated with epigenetic reprogramming of the sperm
exposures rather than chemical exposure could impact ‘epigenetic’
(151,153). Furthermore, paternal nutritional status, drug/toxin expo-
programming. It was recently proposed that the same mechanism
sure and the social environment have been demonstrated to influ-
that provides different tissue-specific identities to identical genes
ence the offspring’s brain and behaviour (via epigenetic
could operate in conferring ‘experience-specific’ identities to genes.
mechanisms) and these effects persist across multiple generations
These mechanisms are proposed to be adaptive/maladaptive physio-
(96). To date, very few published studies have investigated trans-
logical mechanisms that prepare the genome for anticipated life-
generational inheritance of phenotypes induced by prenatal stress
long social and physical environments based on cues received dur-
exposure; however, research in this field is now gathering momen-
ing the perinatal period (156).
tum and our understanding of the mechanisms involved in trans-
generational programming following maternal stress exposure is
Epigenetic mechanisms
likely to increase considerably in the next few years. Epigenetic
reprogramming of the germ cells is a likely mechanism that There are a number of processes acting on different levels of prox-
requires investigation. imity with the DNA molecule itself, which stably alter the accessi-

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 715

bility of the gene to the transcription machinery and define its shown to facilitate demethylation is hydroxylation of the methyl
state of activity. The covalent chemistry of the DNA molecule itself group by ten-eleven translocation enzymes (174). 5-hydroxymethyl-
is altered by DNA methylation (157) and hydroxymethylation (158). cytosine is a poor substrate for DNMT1-mediated maintenance
Histone proteins in chromatin are modulated by a large set of methylation (the copying of methylation from the parental to
chemical modifications such as acetylation (159), methylation (160), daughter strand) (175). During replication, the hydroxyl methylation
phosphorylation ubiquitination (161) and sumoylation (162). Chro- on the parental strand is not copied by DNMT1 to the daughter
matin structure is modulated through altering the positioning of strand and hence the methylation is lost (175). Such a mechanism
nucleosomes. In addition, epigenetic regulation of gene function was recently proposed to be responsible for the loss of DNA meth-
can occur downstream to transcription by involving noncoding and ylation early after fertilisation in the male pronucleus (176). How-
microRNAs, which act on mRNA stability and translation arrest ever, such a mechanism cannot operate in post-mitotic neurones
(163–165). These mechanisms are highly inter-related; for example, because they do not synthesise new DNA. To address this problem
alterations in histone acetylation can result in demethylation and of demethylation in post-mitotic cells, it has recently been proposed
DNA methylation triggers hypoacetylation (166). that DNA hydroxymethylation is removed by ‘passive’ repair mecha-
DNA methylation provides differential identity to DNA during nisms and then an unmethylated cytosine is reintroduced by repair
embryonic development and recent studies have used genome wide DNA synthesis in the absence of DNMT (Fig. 4); thus, the repaired
methods to map the changes in DNA methylation during brain base remains unmethylated (177–181). The identity of the enzymes
development (167). Similarly the ENCODE project has mapped many involved in demethylation in response to ELS are still unknown;
of the tissue specific chromatin modifications; thus, it is well estab- nevertheless, the fact that demethylation occurs in post-mitotic
lished that epigenetic marks participate in cell type specific expres- neurones implies the feasibility of dynamic DNA methylation states
sion. in response to stress.
DNA methylation provides a heritable ‘epigenetic’ mechanism
that confers cell type identity during embryonic development, thus
DNA methylation and the DNA methylation enzymatic
explaining how the same DNA sequence could perform different
machinery
tasks in differentiating cells in a stable and persistent manner. An
In vertebrates, DNA methylation occurs mainly in CG dinucleotide increasing body of data has lent support to the idea that a similar
in DNA (168). Recent evidence suggests that non-CG methylation mechanism occurs in response to postnatal and prenatal stressful
(methylation occurring in CA, CT and CC sequences) is prevalent in experience (Fig. 4). Epigenetic modifications could thus confer upon
the brain mostly in nondividing neurones, where there is no similar genes a different stable functional state in response to
requirement for maintaining a DNA methylation pattern during cell stress and mediate the long-term effects of early-life adversity.
division. Non-CG methylation occurs in promoters in the brain and
might be involved in epigenetic responses of the brain after birth
Involvement of DNA methylation and histone acetylation in
(167,169). DNA methylation is the most proximal epigenetic modifi-
the response to early-life adversity
cation to DNA because it is part of its chemical structure and has
been well established to silence gene expression when occurring in The first evidence for an epigenetic mechanism mediating exposure
50 regulatory regions of genes directly through interference with to ELS was provided by examining the epigenetic effects of differ-
transcription factors binding to their binding recognition element in ences in maternal care in the rat. In this model (discussed above),
DNA (170) or indirectly through attracting methylated DNA binding low licking and grooming by the mother triggers a stable reduction
protein, which in turn recruits chromatin inactivating complexes in expression of NR3C1 in the hippocampus and heightened stress
(171). DNA methylation is catalysed by DNA methyltransferases responsivity in the adult offspring. Examining the mechanisms that
(DNMT) (Fig. 4). DNMT1 is a maintenance DNMT that copies the might mediate such a persistent change in gene expression in
pattern of methylation in the palindromic CG dinucleotide sequence response to maternal behaviour demonstrated that differences in
from the parental strand to the daughter strand during DNA syn- maternal care behaviour resulted in differences in histone acetyla-
thesis in dividing cells. DNMT3a and DNMT3b are de novo methyla- tion DNA methylation at the NR3C1 (GR exon 17 promoter) (133).
tion enzymes because they do not require a methylation template The differences in DNA methylation emerged early after birth in
for methylating DNA. They can therefore methylate non-CG response to maternal care and remained stable into adulthood, pro-
sequences in nondividing cells, which is important in neurones, par- viding a feasible mechanism for environment programming gene
ticularly after birth (167). DNMT3a most probably plays an impor- expression. One of the principal differences between epigenetic and
tant role in this particular brain-specific DNA methylation (169); genetic changes in gene function is that genetic differences are
however, the role of DNMT3a in ELS has not yet been explored. fixed and cannot be changed; however, because DNA methylation
For DNA methylation to play a role in post-mitotic reprogram- and histone acetylation are reversible biochemical reactions (as dis-
ming in response to stress, it must be reversible. Ramchandani cussed above), they should be amenable to pharmacological and
et al. (172) previously proposed that DNA methylation is a bona possibly behavioural interventions. Interestingly, the differences in
fide reversible DNA methylation signal and that a demethylase adult behaviour in response to maternal rearing were reversible in
methylated DNA binding protein 2 (MBD2b) removes the methyl both directions by agents that stimulate DNA methylation such as
group from DNA (173). One mechanism that has recently been the amino acid, methionine (182) and by agents that increase

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
716 S. Maccari et al.

Adversity Neurotransmitters Signalling pathways

Repair: AID, GADD45B, MBD4, BER

Demethylase (MBD2?)

NH2 NH2 NH2


DNMT TET
CH3 CH2OH
N N N

O N O N O N
H SAM SAH H H

Cytosine 5-Methylcytosine 5-Hydroxy-Methylcytosine

Fig. 4. DNA methylation mechanisms targeted by early-life adversity. Cytosine bases in DNA are methylated by DNA methyltransferase (DNMT) enzymes that
catalyse the transfer of a methyl moiety from the methyl donor S-adenosylmethionine (SAM), which is converted to S-adenosylhomocysteine (SAH). Methyla-
tion of cytosine at critical position silences (X) transcription initiation (horizontal green arrow), thus converting an active gene to an inactive gene. The
5-methylcytosine could be further modified by the ten-eleven translocation (TET) oxidases to 5-hydroxymethylcytosine. The epigenetic role of 5-hydroxymethyl-
cytosine is unknown (?). DNA methylation is reversible and could therefore respond to signals from early-life experience. A direct mechanism of demethylation
might involve a demethylase such as MBD2. The widely accepted model is that hydroxymethylated cytosine serves as a substrate for repair-based demethyla-
tion whereby the hydroxymethylated base is removed and replaced with an unmethylated cytosine. This process involves several proteins such as the deami-
nase AID, the repair protein GADD45B and base excision repair enzymes (BER). Demethylation will turn on a silenced gene and thus change its programming.
Early-life experience turns on neuronal pathways, which in turn activate signalling pathways that can potentially act on the multiple methylation and deme-
thylation processes and trigger either DNA methylation or hydroxymethylation and demethylation, thus altering programmes of gene expression.

histone acetylation through inhibition of histone deacetylase with example, exposure of infant rats to stressed caretakers that display
trichostatin A (133). abusive behaviour produces persistent changes in methylation of
A critical question remains: what is the mechanism that mediates the Bdnf gene promoter in the adult prefrontal cortex (185). Simi-
interactions between early-life experience (e.g. maternal care) and larly, ELS in mice causes sustained DNA hypomethylation of an
changes in DNA methylation at particular addresses in the genome? important regulatory region of the arginine vasopressin gene (186).
Weaver et al. (183) proposed that maternal care leads to activation More recently, it has been shown that a post-traumatic stress dis-
of a transcription factor, nerve growth factor inducible gene-A order risk allele of the proximal regulator of GR, FKBP5, is demethy-
(NGFI-A) in response to serotonin receptor activation, which in turn lated in peripheral lymphocytes in children who were exposed to
targets the NR3C1 promoter for DNA demethylation. This provides trauma early in life (187). This study elegantly demonstrates a
a feasible paradigm of a conduit leading from exposure to stress to methylation-dependent ‘gene–environment’ interaction (187).
changes in DNA methylation at particular sites in the genome. Initial studies exploring an epigenetic mechanism for ELS focused
These results have been translated to humans. By examining hip- on candidate genes and on specific brain regions where well estab-
pocampi from humans who were exposed to child abuse and com- lished physiological stress regulation circuitries are positioned.
paring the DNA methylation at the same NR3C1 promoter, However, it is clear, as discussed above, that ELS is associated with
McGowan et al. (184) showed that child abuse is associated with multiple adult phenotypes. Therefore, it has been hypothesised that
hypermethylation of several sites, including the NGFI-A recognition the changes in DNA methylation triggered by ELS should not be
element that was implicated in targeting demethylation in response limited to several candidate genes and instead must be system-
to maternal care licking and grooming in rats. The changes in DNA wide involving multiple physiological systems. Furthermore, it is
methylation are associated with differences in expression of the proposed that an integrated response of the methylome to physical,
NR3C1 receptor, linking child abuse and hippocampal regulation of social and biosphere cues occurs early in life as an adaptation of
stress responses (184). the genome to anticipated life-long experiences (188). These adap-
Several other studies support the hypothesis that ELS results in tations could become maladaptive if social and other environmental
particular changes in DNA methylation in candidate genes. For contexts are altered later in life (188).

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 717

Consistent with this hypothesis, McGowan et al. (189) have particularly susceptible to early-life programming and this is mani-
shown that child abuse is associated with an alteration in the state fested as stress hyper-reactivity, increased susceptibility to affective
of methylation of the ubiquitous promoters of ribosomal RNA disorders (e.g. anxiety and depression) and cognitive deficits. In the
(rRNA) genes, which are critical for general protein synthesis but case of prenatal stress, the mechanisms underlying how the effects
were not previously implicated in responses to ELS. This change in are transmitted from the mother to the offspring are not fully
DNA methylation is associated with differences in the expression of understood but, given that stress in gestation can negatively affect
rRNA (189). Similarly, in rats, a transcriptome analysis of the hippo- subsequent maternal care, and, moreover, maternally-deprived off-
campus from adult offspring of high and low licking/grooming spring display similar phenotypes to prenatally stressed animals,
dams revealed differences in hundreds of transcripts (190). Broad disrupted maternal care postpartum is one possibility.
changes in DNA methylation, histone acetylation and transcription Studies aimed at reversing the negative effects of early-life
covering a few megabytes around the NR3C1 locus in the hippo- adversity by exploiting findings from what we understand about
campus were revealed in adult rats that received differential mater- the neurobiological and epigenetic mechanisms underpinning some
nal care postnatally (191) and in humans that were abused as of the phenotypes observed in programmed offspring should pro-
children (192). vide insights for developing new therapeutic strategies for the
Recent studies examining the impact of exposure of guinea pig treatment of mood disorders and cognitive decline. Moreover, the
foetuses to glucocorticoids prenatally showed changes in overall finding that the negative phenotypes arising from early-life adver-
DNA methylation in the adult offspring in several tissues that per- sity may potentially be transmitted to subsequent generations has
sisted for two generations, as well as genome wide changes in wide-ranging implications for disease susceptibility in humans and
DNA methylation states, histone acetylation and transcription in further research in this area is warranted.
exposed foetal hippocampi (193,194). Although data are limited, evidence is emerging to support the
A critical question for the study of behavioural epigenetics, par- hypothesis that DNA methylation is a potential mediator of the
ticularly in humans, is whether DNA methylation associated with long-term response to early-life adversity. These responses are pro-
exposure to ELS is limited to the brain or whether they are system posed to be an adaptive response of the genome to anticipated
wide (188). This obviously has important implications because the integrated stressful environment that is predicted by early-life cues.
primary target of stress regulation, the brain, is not accessible in The response is not limited to a few candidate genes; it involves
living humans or animals. Provencal et al. (195) examined the several gene pathways and occurs in both central and peripheral
impact of differential rearing in nonhuman primates (rhesus maca- tissues. The fact that changes in DNA methylation also occur in
ques) in parallel with DNA methylation in the prefrontal cortex and peripheral tissues indicates that it might be possible to study the
peripheral T cells. Differences in DNA methylation were detected in immune facet of stress in living humans in longitudinal cohorts, as
both tissues; however, it is important to note that the changes in well as in response to behavioural and pharmacological interven-
DNA methylation are not surrogate markers of the prefrontal cor- tions.
tex, although they most probably reflect the response of the
immune system to ELS. Nevertheless, several differences are com-
Acknowledgements
mon in both tissues.
The idea that the epigenomic response to ELS is not limited to Research performed in the laboratory of SM and SMF is funded by the Fon-
the brain and could be studied in peripheral lymphocytes has dation de France and the International Associated Laboratory for prenatal
stress and neurodegenerative diseases (LIA-PSND). Research by HJK is finan-
gained support from several other tissues. The early-life trauma
cially supported by the Netherlands Organization for Scientific Research
associated demethylation of FKBP5 is detected in peripheral white
(NWO), Cognitive Science Center Amsterdam, Internationale Stitchting Alz-
cells (187) and the same group have recently shown a DNA meth- heimer Onderzoek (ISAO). Studies performed in MS’s laboratory are sup-
ylation signature of early-life adversity in peripheral blood cells ported by the Canadian Institute for Health Research and the Sackler
(196). Moreover, Borghol et al. (197) have demonstrated a broad program in Epigenetics and Psychobiology. PJB receives funding from the
DNA methylation signature associated with socioeconomic position- Biotechnology and Biological Sciences Research Council (BBSRC) and the
ing in whole blood DNA from 45-year-old adults. Other studies British Society for Neuroendocrinology.
have correlated DNA methylation of the NR3C1 gene in DNA from
neonatal cord blood mononuclear cells with maternal depression Received 16 January 2014,
(198) and childhood adversity and maltreatment (199–201). revised 9 July 2014,
Together, these findings give rise to the potential for longitudinal accepted 10 July 2014
studies investigating possible environmental or pharmacological
interventions. References
1 Brunton PJ, Russell JA. Prenatal social stress in the rat programmes
Summary and outlook neuroendocrine and behavioural responses to stress in the adult off-
spring: sex specific effects. J Neuroendocrinol 2010; 22: 258–271.
In summary, adverse experiences occurring in early life, be it pre- 2 Brunton PJ, Sullivan KM, Kerrigan D, Russell JA, Seckl JR, Drake AJ.
or postnatally, can have life-long detrimental consequences, which Sex-specific effects of prenatal stress on glucose homoeostasis and
are associated with an increased propensity for disease. The brain is peripheral metabolism in rats. J Endocrinol 2013; 217: 161–173.

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
718 S. Maccari et al.

3 Lesage J, Del-Favero F, Leonhardt M, Louvart H, Maccari S, Vieau D, spring: potential relevance to schizophrenia. Behav Brain Res 2005;
Darnaudery M. Prenatal stress induces intrauterine growth restriction 156: 251–261.
and programmes glucose intolerance and feeding behaviour distur- 23 Mueller BR, Bale TL. Sex-specific programming of offspring emotional-
bances in the aged rat. J Endocrinol 2004; 181: 291–296. ity after stress early in pregnancy. J Neurosci 2008; 28: 9055–9065.
4 Maccari S, Darnaudery M, Morley-Fletcher S, Zuena AR, Cinque C, Van 24 Lee PR, Brady DL, Shapiro RA, Dorsa DM, Koenig JI. Prenatal stress
Reeth O. Prenatal stress and long-term consequences: implications of generates deficits in rat social behavior: reversal by oxytocin. Brain Res
glucocorticoid hormones. Neurosci Biobehav Rev 2003; 27: 119–127. 2007; 1156: 152–167.
5 Gluckman PD, Hanson MA. Developmental origins of disease paradigm: 25 Brunton PJ. Effects of maternal exposure to social stress during preg-
a mechanistic and evolutionary perspective. Pediatr Res 2004; 56: nancy: consequences for mother and offspring. Reproduction 2013;
311–317. 146: R175–R189.
6 Seckl JR, Holmes MC. Mechanisms of disease: glucocorticoids, their 26 Maccari S, Piazza PV, Kabbaj M, Barbazanges A, Simon H, Le Moal M.
placental metabolism and fetal ‘programming’ of adult pathophysiol- Adoption reverses the long-term impairment in glucocorticoid feedback
ogy. Nat Clin Pract Endocrinol Metab 2007; 3: 479–488. induced by prenatal stress. J Neurosci 1995; 15: 110–116.
7 Barker DJ. Intrauterine programming of adult disease. Mol Med Today 27 Morley-Fletcher S, Darnaudery M, Koehl M, Casolini P, Van Reeth O,
1995; 1: 418–423. Maccari S. Prenatal stress in rats predicts immobility behavior in the
8 Barker DJ, Bagby SP, Hanson MA. Mechanisms of disease: in utero forced swim test. Effects of a chronic treatment with tianeptine. Brain
programming in the pathogenesis of hypertension. Nat Clin Pract Res 2003; 989: 246–251.
Nephrol 2006; 2: 700–707. 28 Koehl M, Darnaudery M, Dulluc J, Van Reeth O, Le Moal M, Maccari S.
9 Meaney MJ, Szyf M, Seckl JR. Epigenetic mechanisms of perinatal pro- Prenatal stress alters circadian activity of hypothalamo-pituitary-adre-
gramming of hypothalamic-pituitary-adrenal function and health. nal axis and hippocampal corticosteroid receptors in adult rats of both
Trends Mol Med 2007; 13: 269–277. gender. J Neurobiol 1999; 40: 302–315.
10 Darnaudery M, Maccari S. Epigenetic programming of the stress 29 Sapolsky RM, Meaney MJ. Maturation of the adrenocortical stress
response in male and female rats by prenatal restraint stress. Brain response: neuroendocrine control mechanisms and the stress hypore-
Res Rev 2008; 57: 571–585. sponsive period. Brain Res 1986; 396: 64–76.
11 Weinstock M. The long-term behavioural consequences of prenatal 30 Vallee M, MacCari S, Dellu F, Simon H, Le Moal M, Mayo W. Long-term
stress. Neurosci Biobehav Rev 2008; 32: 1073–1086. effects of prenatal stress and postnatal handling on age-related gluco-
12 Lupien SJ, McEwen BS, Gunnar MR, Heim C. Effects of stress through- corticoid secretion and cognitive performance: a longitudinal study in
out the lifespan on the brain, behaviour and cognition. Nat Rev Neuro- the rat. Eur J Neurosci 1999; 11: 2906–2916.
sci 2009; 10: 434–445. 31 Koehl M, Barbazanges A, Le Moal M, Maccari S. Prenatal stress induces
13 Shonkoff JP, Boyce WT, McEwen BS. Neuroscience, molecular biology, a phase advance of circadian corticosterone rhythm in adult rats which
and the childhood roots of health disparities: building a new frame- is prevented by postnatal stress. Brain Res 1997; 759: 317–320.
work for health promotion and disease prevention. JAMA 2009; 301: 32 Dugovic C, Maccari S, Weibel L, Turek FW, Van Reeth O. High cortico-
2252–2259. sterone levels in prenatally stressed rats predict persistent paradoxical
14 Krishnan V, Nestler EJ. The molecular neurobiology of depression. sleep alterations. J Neurosci 1999; 19: 8656–8664.
Nature 2008; 455: 894–902. 33 Mairesse J, Silletti V, Laloux C, Zuena AR, Giovine A, Consolazione M,
15 McEwen BS. The ever-changing brain: cellular and molecular mecha- van Camp G, Malagodi M, Gaetani S, Cianci S, Catalani A, Mennuni G,
nisms for the effects of stressful experiences. Dev Neurobiol 2012; 72: Mazzetta A, van Reeth O, Gabriel C, Mocaer E, Nicoletti F, Morley-
878–890. Fletcher S, Maccari S. Chronic agomelatine treatment corrects the
16 Henry C, Kabbaj M, Simon H, Le Moal M, Maccari S. Prenatal stress abnormalities in the circadian rhythm of motor activity and sleep/wake
increases the hypothalamo-pituitary-adrenal axis response in young cycle induced by prenatal restraint stress in adult rats. Int J Neuropsy-
and adult rats. J Neuroendocrinol 1994; 6: 341–345. chopharmacol 2013; 16: 323–338.
17 Fride E, Weinstock M. The effects of prenatal exposure to predictable 34 Maccari S, Van Reeth O. Circadian rhythms, effects of prenatal stress
or unpredictable stress on early development in the rat. Dev Psychobiol in rodents. In: Fink G, ed. Encyclopedia of Stress. 2nd edn. Amsterdam:
1984; 17: 651–660. Elsevier, 2007; 505–508.
18 Kapoor A, Matthews SG. Short periods of prenatal stress affect growth, 35 Laloux C, Mairesse J, Van Camp G, Giovine A, Branchi I, Bouret S, Mor-
behaviour and hypothalamo-pituitary-adrenal axis activity in male gui- ley-Fletcher S, Bergonzelli G, Malagodi M, Gradini R, Nicoletti F, Dar-
nea pig offspring. J Physiol 2005; 566: 967–977. naudery M, Maccari S. Anxiety- like behaviour and associated
19 Fan JM, Chen XQ, Jin H, Du JZ. Gestational hypoxia alone or combined neurochemical and endocrinological alterations in male pups exposed
with restraint sensitizes the hypothalamic-pituitary-adrenal axis and to prenatal stress. Psychoneuroendocrinology 2012; 37: 1646–1658.
induces anxiety-like behavior in adult male rat offspring. Neuroscience 36 Morley-Fletcher S, Rea M, Maccari S, Laviola G. Environmental enrich-
2009; 159: 1363–1373. ment during adolescence reverses the effects of prenatal stress on play
20 Paris JJ, Brunton PJ, Russell JA, Frye CA. Immune stress in late preg- behaviour and HPA axis reactivity in rats. Eur J Neurosci 2003; 18:
nant rats decreases length of gestation, fecundity, and alters later cog- 3367–3374.
nitive and affective behaviour of surviving pre-adolescent offspring. 37 Vallee M, Mayo W, Dellu F, Le Moal M, Simon H, Maccari S. Prenatal
Stress 2011; 14: 652–664. stress induces high anxiety and postnatal handling induces low anxiety
21 Bosch OJ, Musch W, Bredewold R, Slattery DA, Neumann ID. Prenatal in adult offspring: correlation with stress-induced corticosterone secre-
stress increases HPA axis activity and impairs maternal care in lactat- tion. J Neurosci 1997; 17: 2626–2636.
ing female offspring: implications for postpartum mood disorder. 38 Morley-Fletcher S, Darnaudery M, Mocaer E, Froger N, Lanfumey L, La-
Psychoneuroendocrinology 2007; 32: 267–278. viola G, Casolini P, Zuena AR, Marzano L, Hamon M, Maccari S. Chronic
22 Koenig JI, Elmer GI, Shepard PD, Lee PR, Mayo C, Joy B, Hercher E, treatment with imipramine reverses immobility behaviour, hippocampal
Brady DL. Prenatal exposure to a repeated variable stress paradigm corticosteroid receptors and cortical 5- HT(1A) receptor mRNA in pre-
elicits behavioral and neuroendocrinological changes in the adult off- natally stressed rats. Neuropharmacology 2004; 47: 841–847.

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 719

39 Morley-Fletcher S, Mairesse J, Soumier A, Banasr M, Fagioli F, Gabriel robiological alterations. Psychoneuroendocrinology 2007; 32(Suppl. 1):
C, Mocaer E, Daszuta A, McEwen B, Nicoletti F, Maccari S. Chronic ago- S10–S15.
melatine treatment corrects behavioral, cellular, and biochemical 56 Marrocco J, Reynaert ML, Gatta E, Gabriel C, Moca€er E, Di Prisco S,
abnormalities induced by prenatal stress in rats. Psychopharmacology Merega E, Anna Pittaluga A, Nicoletti F, Maccari S, Morley-Fletcher S,
2011; 217: 301–313. Jer^ome Mairesse J. The effects of antidepressant treatment in prena-
40 Van Waes V, Darnaudery M, Marrocco J, Gruber SH, Talavera E, tally stressed rats support the glutamatergic hypothesis of stress-
Mairesse J, Van Camp G, Casolla B, Nicoletti F, Mathe AA, Maccari S, related disorders. J Neurosci 2014; 34: 2015–20240.
Morley-Fletcher S. Impact of early life stress on alcohol consumption 57 Dunn AJ, Berridge CW. Physiological and behavioral responses to corti-
and on the short- and long-term responses to alcohol in adolescent cotropin-releasing factor administration: is CRF a mediator of anxiety
female rats. Behav Brain Res 2011; 221: 43–49. or stress responses? Brain Res Rev 1990; 15: 71–100.
41 Zuena AR, Mairesse J, Casolini P, Cinque C, Alema GS, Morley-Fletcher 58 Schulkin J, Gold PW, McEwen BS. Induction of corticotropin-releasing
S, Chiodi V, Spagnoli LG, Gradini R, Catalani A, Nicoletti F, Maccari S. hormone gene expression by glucocorticoids: implication for under-
Prenatal restraint stress generates two distinct behavioral and neuro- standing the states of fear and anxiety and allostatic load. Psychoneu-
chemical profiles in male and female rats. PLoS ONE 2008; 3: e2170. roendocrinology 1998; 23: 219–243.
42 Coe CL, Kramer M, Czeh B, Gould E, Reeves AJ, Kirschbaum C, Fuchs E. 59 Ward HE, Johnson EA, Salm AK, Birkle DL. Effects of prenatal stress on
Prenatal stress diminishes neurogenesis in the dentate gyrus of juve- defensive withdrawal behavior and corticotropin releasing factor sys-
nile rhesus monkeys. Biol Psychiatry 2003; 54: 1025–1034. tems in rat brain. Physiol Behav 2000; 70: 359–366.
43 Son GH, Geum D, Chung S, Kim EJ, Jo JH, Kim CM, Lee KH, Kim H, Choi 60 Makino S, Gold PW, Schulkin J. Corticosterone effects on corticotropin-
S, Kim HT, Lee CJ, Kim K. Maternal stress produces learning deficits releasing hormone mRNA in the central nucleus of the amygdala and
associated with impairment of NMDA receptor-mediated synaptic plas- the parvocellular region of the paraventricular nucleus of the hypo-
ticity. J Neurosci 2006; 26: 3309–3318. thalamus. Brain Res 1994; 640: 105–112.
44 Mueller BR, Bale TL. Early prenatal stress impact on coping strategies 61 Brunton PJ, Donadio MV, Russell JA. Sex differences in prenatally pro-
and learning performance is sex dependent. Physiol Behav 2007; 91: grammed anxiety behaviour in rats: differential corticotropin-releasing
55–65. hormone receptor mRNA expression in the amygdaloid complex. Stress
45 Darnaudery M, Perez-Martin M, Belizaire G, Maccari S, Garcia-Segura 2011; 14: 634–643.
LM. Insulin-like growth factor 1 reduces age-related disorders induced 62 Bale TL, Vale WW. CRF and CRF receptors: role in stress responsivity
by prenatal stress in female rats. Neurobiol Aging 2006; 27: 119–127. and other behaviors. Annu Rev Pharmacol Toxicol 2004; 44: 525–557.
46 Van Waes V, Enache M, Zuena A, Mairesse J, Nicoletti F, Vinner E, Lher- 63 Wang X, Meng FS, Liu ZY, Fan JM, Hao K, Chen XQ, Du JZ. Gestational
mitte M, Maccari S, Darnaudery M. Ethanol attenuates spatial memory hypoxia induces sex-differential methylation of Crhr1 linked to anxi-
deficits and increases mGlu1a receptor expression in the hippocampus ety-like behavior. Mol Neurobiol 2013; 48: 554–555.
of rats exposed to prenatal stress. Alcohol Clin Exp Res 2009; 33: 64 Zohar I, Weinstock M. Differential effect of prenatal stress on the
1346–1354. expression of corticotrophin-releasing hormone and its receptors in the
47 Lemaire V, Koehl M, Le Moal M, Abrous DN. Prenatal stress produces hypothalamus and amygdala in male and female rats. J Neuroendocri-
learning deficits associated with an inhibition of neurogenesis in the nol 2011; 23: 320–328.
hippocampus. Proc Natl Acad Sci USA 2000; 97: 11032–11037. 65 Wang XD, Labermaier C, Holsboer F, Wurst W, Deussing JM, Muller MB,
48 Bosch OJ, Kromer SA, Neumann ID. Prenatal stress: opposite effects on Schmidt MV. Early-life stress-induced anxiety-related behavior in adult
anxiety and hypothalamic expression of vasopressin and corticotropin- mice partially requires forebrain corticotropin-releasing hormone recep-
releasing hormone in rats selectively bred for high and low anxiety. tor 1. Eur J Neurosci 2012; 36: 2360–2367.
Eur J Neurosci 2006; 23: 541–551. 66 Welberg LAM, Seckl JR. Prenatal stress, glucocorticoids and the pro-
49 Lucassen PJ, Bosch OJ, Jousma E, Kromer SA, Andrew R, Seckl JR, gramming of the brain. J Neuroendocrinol 2001; 13: 113–128.
Neumann ID. Prenatal stress reduces postnatal neurogenesis in rats 67 Seckl JR. Glucocorticoids, developmental ‘programming’ and the risk of
selectively bred for high, but not low, anxiety: possible key role of pla- affective dysfunction. Prog Brain Res 2008; 167: 17–34.
cental 11beta-hydroxysteroid dehydrogenase type 2. Eur J Neurosci 68 Welberg LAM, Seckl JR, Holmes MC. Prenatal glucocorticoid program-
2009; 29: 97–103. ming of brain corticosteroid receptors and corticotrophin-releasing
50 Szyf M, Weaver IC, Champagne FA, Diorio J, Meaney MJ. Maternal pro- hormone: possible implications for behaviour. Neuroscience 2001; 104:
gramming of steroid receptor expression and phenotype through DNA 71–79.
methylation in the rat. Front Neuroendocrinol 2005; 26: 139–162. 69 Barbazanges A, Piazza PV, Le Moal M, Maccari S. Maternal glucocorti-
51 Ongur D, Jensen JE, Prescot AP, Stork C, Lundy M, Cohen BM, Renshaw coid secretion mediates long-term effects of prenatal stress. J Neurosci
PF. Abnormal glutamatergic neurotransmission and neuronal-glial 1996; 16: 3943–3949.
interactions in acute mania. Biol Psychiatry 2008; 64: 718–726. 70 Benediktsson R, Calder AA, Edwards CR, Seckl JR. Placental 11 beta-hy-
52 Chen G, Henter ID, Manji HK. Presynaptic glutamatergic dysfunction in droxysteroid dehydrogenase: a key regulator of fetal glucocorticoid
bipolar disorder. Biol Psychiatry 2010; 67: 1007–1009. exposure. Clin Endocrinol (Oxf) 1997; 46: 161–166.
53 Popoli M, Yan Z, McEwen BS, Sanacora G. The stressed synapse: the 71 Welberg LA, Thrivikraman KV, Plotsky PM. Chronic maternal stress
impact of stress and glucocorticoids on glutamate transmission. Nat inhibits the capacity to up-regulate placental 11beta-hydroxysteroid
Rev Neurosci 2012; 13: 22–37. dehydrogenase type 2 activity. J Endocrinol 2005; 186: R7–R12.
54 Marrocco J, Mairesse J, Ngomba RT, Silletti V, Van Camp G, Bouwalerh 72 Mairesse J, Lesage J, Breton C, Breant B, Hahn T, Darnaudery M, Dick-
H, Summa M, Pittaluga A, Nicoletti F, Maccari S, Morley-Fletcher S. son SL, Seckl J, Blondeau B, Vieau D, Maccari S, Viltart O. Maternal
Anxiety-like behavior of prenatally stressed rats is associated with a stress alters endocrine function of the feto-placental unit in rats. Am J
selective reduction of glutamate release in the ventral hippocampus. J Physiol Endocrinol Metab 2007; 292: E1526–E1533.
Neurosci 2012; 32: 17143–17154. 73 Brunton PJ, Russell JA, Douglas AJ. Adaptive responses of the maternal
55 Maccari S, Morley-Fletcher S. Effects of prenatal restraint stress on the hypothalamic-pituitary-adrenal axis during pregnancy and lactation. J
hypothalamus-pituitary-adrenal axis and related behavioural and neu- Neuroendocrinol 2008; 20: 764–776.

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
720 S. Maccari et al.

74 Atkinson HC, Waddell BJ. The hypothalamic-pituitary-adrenal axis in 91 Smith JW, Seckl JR, Evans AT, Costall B, Smythe JW. Gestational stress
rat pregnancy and lactation: circadian variation and interrelationship induces post-partum depression-like behaviour and alters maternal
of plasma adrenocorticotropin and corticosterone. Endocrinology 1995; care in rats. Psychoneuroendocrinology 2004; 29: 227–244.
136: 512–520. 92 Hillerer KM, Reber SO, Neumann ID, Slattery DA. Exposure to chronic
75 Ohkawa T, Rohde W, Takeshita S, Dorner G, Arai K, Okinaga S. Effect of pregnancy stress reverses peripartum-associated adaptations: implica-
an acute maternal stress on the fetal hypothalamo-pituitary-adrenal tions for postpartum anxiety and mood disorders. Endocrinology 2011;
system in late gestational life of the rat. Exp Clin Endocrinol 1991; 98: 152: 3930–3940.
123–129. 93 Lukas M, Bredewold R, Neumann ID, Veenema AH. Maternal separation
76 Fujioka T, Fujioka A, Endoh H, Sakata Y, Furukawa S, Nakamura S. interferes with developmental changes in brain vasopressin and oxyto-
Materno-fetal coordination of stress-induced Fos expression in the cin receptor binding in male rats. Neuropharmacology 2010; 58: 78–
hypothalamic paraventricular nucleus during pregnancy. Neuroscience 87.
2003; 118: 409–415. 94 Kenkel WM, Yee JR, Carter CS. Is oxytocin a maternal-fetal signaling
77 Russell JA, Douglas AJ, Brunton PJ. Reduced hypothalamo-pituitary- molecule at birth? Implications for development J Neuroendocrinol
adrenal axis stress responses in late pregnancy: central opioid inhibi- 2014; 26: 739–749.
tion and noradrenergic mechanisms. Ann NY Acad Sci 2008; 1148: 95 Lucassen PJ, Naninck EF, van Goudoever JB, Fitzsimons C, Joels zM,
428–438. Korosi A. Perinatal programming of adult hippocampal structure and
78 Vaha-Eskeli KK, Erkkola RU, Scheinin M, Seppanen A. Effects of short- function; emerging roles of stress, nutrition and epigenetics. Trends
term thermal stress on plasma catecholamine concentrations and Neurosci 2013; 36: 621–631.
plasma renin activity in pregnant and nonpregnant women. Am J 96 Braun K, Champagne FA. Paternal influences on offspring development:
Obstet Gynecol 1992; 167: 785–789. behavioural and epigenetic pathways. J Neuroendocrinol 2014; 26:
79 Roelfsema V, Gunn AJ, Fraser M, Quaedackers JS, Bennet L. Cortisol 697–706.
and ACTH responses to severe asphyxia in preterm fetal sheep. Exp 97 De Kloet ER, Rosenfeld P, Van Eekelen JA, Sutanto W, Levine S. Stress,
Physiol 2005; 90: 545–555. glucocorticoids and development. Prog Brain Res 1988; 73: 101–120.
80 Edwards LJ, McMillen IC. Impact of maternal undernutrition during the 98 Stanton ME, Gutierrez YR, Levine S. Maternal deprivation potentiates
periconceptional period, fetal number, and fetal sex on the develop- pituitary- adrenal stress responses in infant rats. Behav Neurosci 1988;
ment of the hypothalamo-pituitary adrenal axis in sheep during late 102: 692–700.
gestation. Biol Reprod 2002; 66: 1562–1569. 99 Levine S. The ontogeny of the hypothalamic-pituitary-adrenal axis. The
81 Gu W, Jones CT. The effect of elevation of maternal plasma catechol- influence of maternal factors. Ann NY Acad Sci 1994; 746: 275–293.
amines on the fetus and placenta of the pregnant sheep. J Dev Physiol 100 Schmidt M, Okimoto DK, Dent GW, Gordon MK, Levine S. Maternal reg-
1986; 8: 173–186. ulation of the hypothalamic-pituitary-adrenal axis in the 20-day-old
82 Lingas RI, Matthews SG. A short period of maternal nutrient restriction rat: consequences of laboratory weaning. J Neuroendocrinol 2002; 14:
in late gestation modifies pituitary-adrenal function in adult guinea 450–457.
pig offspring. Neuroendocrinology 2001; 73: 302–311. 101 Schmidt MV, Oitzl MS, Levine S, de Kloet ER. The HPA system during
83 Nunez H, Ruiz S, Soto-Moyano R, Navarrete M, Valladares L, White A, the postnatal development of CD1 mice and the effects of maternal
Perez H. Fetal undernutrition induces overexpression of CRH mRNA deprivation. Brain Res Dev Brain Res 2002; 139: 39–49.
and CRH protein in hypothalamus and increases CRH and corticoste- 102 Rosenfeld P, Suchecki D, Levine S. Multifactorial regulation of the
rone in plasma during postnatal life in the rat. Neurosci Lett 2008; hypothalamic- pituitary-adrenal axis during development. Neurosci
448: 115–119. Biobehav Rev 1992; 16: 553–568.
84 Knipp GT, Audus KL, Soares MJ. Nutrient transport across the placenta. 103 Plotsky PM, Meaney MJ. Early, postnatal experience alters hypothalamic
Adv Drug Deliv Rev 1999; 38: 41–58. corticotropin-releasing factor (CRF) mRNA, median eminence CRF con-
85 Korgun ET, Demir R, Hammer A, Dohr G, Desoye G, Skofitsch G, Hahn tent and stress- induced release in adult rats. Brain Res Mol Brain Res
T. Glucose transporter expression in rat embryo and uterus during de- 1993; 18: 195–200.
cidualization, implantation, and early postimplantation. Biol Reprod 104 Ogawa T, Mikuni M, Kuroda Y, Muneoka K, Mori KJ, Takahashi K. Peri-
2001; 65: 1364–1370. odic maternal deprivation alters stress response in adult offspring:
86 Howerton CL, Morgan CP, Fischer DB, Bale TL. O-GlcNAc transferase potentiates the negative feedback regulation of restraint stress-
(OGT) as a placental biomarker of maternal stress and reprogramming induced adrenocortical response and reduces the frequencies of open
of CNS gene transcription in development. Proc Natl Acad Sci USA field-induced behaviors. Pharmacol Biochem Behav 1994; 49:
2013; 110: 5169–5174. 961–967.
87 Li MD, Ruan HB, Singh JP, Zhao L, Zhao T, Azarhoush S, Wu J, 105 Vazquez DM, Van Oers H, Levine S, Akil H. Regulation of glucocorticoid
Evans RM, Yang X. O-GlcNAc transferase is involved in glucocorticoid and mineralocorticoid receptor mRNAs in the hippocampus of the
receptor-mediated transrepression. J Biol Chem 2012; 287: maternally deprived infant rat. Brain Res 1996; 731: 79–90.
12904–12912. 106 Liu D, Caldji C, Sharma S, Plotsky PM, Meaney MJ. Influence of neona-
88 Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, Freedman A, Sharma tal rearing conditions on stress-induced adrenocorticotropin responses
S, Pearson D, Plotsky PM, Meaney MJ. Maternal care, hippocampal glu- and norepinepherine release in the hypothalamic paraventricular
cocorticoid receptors, and hypothalamic-pituitary-adrenal responses to nucleus. J Neuroendocrinol 2000; 12: 5–12.
stress. Science 1997; 277: 1659–1662. 107 Lehmann J, Russig H, Feldon J, Pryce CR. Effect of a single maternal
89 Champagne F, Meaney MJ. Like mother, like daughter: evidence for separation at different pup ages on the corticosterone stress response
non-genomic transmission of parental behaviour and stress responsivi- in adult and aged rats. Pharmacol Biochem Behav 2002; 73: 141–145.
ty. Prog Brain Res 2001; 133: 287–302. 108 Ladd CO, Huot RL, Thrivikraman KV, Nemeroff CB, Plotsky PM. Long-
90 Moore CL, Power KL. Prenatal stress affects mother-infant interaction term adaptations in glucocorticoid receptor and mineralocorticoid
in Norway rats. Dev Psychobiol 1986; 19: 235–245. receptor mRNA and negative feedback on the hypothalamo-pituitary-

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 721

adrenal axis following neonatal maternal separation. Biol Psychiatry 126 Wang XD, Rammes G, Kraev I, Wolf M, Liebl C, Scharf SH, Rice CJ,
2004; 55: 367–375. Wurst W, Holsboer F, Deussing JM, Baram TZ, Stewart MG, Muller MB,
109 Haller J, Harold G, Sandi C, Neumann ID. Effects of adverse early-life Schmidt MV. Forebrain CRF(1) modulates early-life stress-programmed
events on aggression and anti-social behaviours in animals and cognitive deficits. J Neurosci 2011; 31: 13625–13634.
humans. J Neuroendocrinol 2014; 26: 724–738. 127 Ivy AS, Rex CS, Chen Y, Dube C, Maras PM, Grigoriadis DE, Gall CM,
110 Uysal N, Ozdemir D, Dayi A, Yalaz G, Baltaci AK, Bediz CS. Effects of Lynch G, Baram TZ. Hippocampal dysfunction and cognitive impair-
maternal deprivation on melatonin production and cognition in adoles- ments provoked by chronic early- life stress involve excessive activa-
cent male and female rats. Neuro Endocrinol Lett 2005; 26: 555–560. tion of CRH receptors. J Neurosci 2010; 30: 13005–13015.
111 Aisa B, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Cognitive impair- 128 Barnett SA, Burn J. Early stimulation and maternal behaviour. Nature
ment associated to HPA axis hyperactivity after maternal separation in 1967; 213: 150–152.
rats. Psychoneuroendocrinology 2007; 32: 256–266. 129 Smotherman WP, Brown CP, Levine S. Maternal responsiveness follow-
112 Oomen CA, Soeters H, Audureau N, Vermunt L, van Hasselt FN, Mand- ing differential pup treatment and mother-pup interactions. Horm
ers EM, Joels M, Lucassen PJ, Krugers H. Severe early life stress ham- Behav 1977; 8: 242–253.
pers spatial learning and neurogenesis, but improves hippocampal 130 Pryce CR, Bettschen D, Feldon J. Comparison of the effects of early
synaptic plasticity and emotional learning under high-stress conditions handling and early deprivation on maternal care in the rat. Dev
in adulthood. J Neurosci 2010; 30: 6635–6645. Psychobiol 2001; 38: 239–251.
113 McIntosh J, Anisman H, Merali Z. Short- and long-periods of neonatal 131 Myers MM, Brunelli SA, Squire JM, Shindeldecker RD, Hofer MA. Mater-
maternal separation differentially affect anxiety and feeding in adult nal behavior of SHR rats and its relationship to offspring blood pres-
rats: gender-dependent effects. Brain Res Dev Brain Res 1999; 113: sures. Dev Psychobiol 1989; 22: 29–53.
97–106. 132 Champagne FA, Francis DD, Mar A, Meaney MJ. Variations in maternal
114 Caldji C, Francis D, Sharma S, Plotsky PM, Meaney MJ. The effects of care in the rat as a mediating influence for the effects of environment
early rearing environment on the development of GABAA and central on development. Physiol Behav 2003; 79: 359–371.
benzodiazepine receptor levels and novelty-induced fearfulness in the 133 Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl
rat. Neuropsychopharmacology 2000; 22: 219–229. JR, Dymov S, Szyf M, Meaney MJ. Epigenetic programming by maternal
115 Kalinichev M, Easterling KW, Plotsky PM, Holtzman SG. Long-lasting behavior. Nat Neurosci 2004; 7: 847–854.
changes in stress- induced corticosterone response and anxiety-like 134 Liu D, Diorio J, Day JC, Francis DD, Meaney MJ. Maternal care, hippo-
behaviors as a consequence of neonatal maternal separation in Long- campal synaptogenesis and cognitive development in rats. Nat Neuro-
Evans rats. Pharmacol Biochem Behav 2002; 73: 131–140. sci 2000; 3: 799–806.
116 Huot RL, Plotsky PM, Lenox RH, McNamara RK. Neonatal maternal sep- 135 Champagne DL, Bagot RC, van Hasselt F, Ramakers G, Meaney MJ, de
aration reduces hippocampal mossy fiber density in adult Long Evans Kloet ER, Joels M, Krugers H. Maternal care and hippocampal plasticity:
rats. Brain Res 2002; 950: 52–63. evidence for experience- dependent structural plasticity, altered synap-
117 Mirescu C, Peters JD, Gould E. Early life experience alters response of tic functioning, and differential responsiveness to glucocorticoids and
adult neurogenesis to stress. Nat Neurosci 2004; 7: 841–846. stress. J Neurosci 2008; 28: 6037–6045.
118 Aisa B, Elizalde N, Tordera R, Lasheras B, Del Rio J, Ramirez MJ. Effects 136 Bredy TW, Grant RJ, Champagne DL, Meaney MJ. Maternal care influ-
of neonatal stress on markers of synaptic plasticity in the hippocam- ences neuronal survival in the hippocampus of the rat. Eur J Neurosci
pus: implications for spatial memory. Hippocampus 2009; 19: 2003; 18: 2903–2909.
1222–1231. 137 Bagot RC, van Hasselt FN, Champagne DL, Meaney MJ, Krugers HJ,
119 Oomen CA, Girardi CE, Cahyadi R, Verbeek EC, Krugers H, Joels M, Joels M. Maternal care determines rapid effects of stress mediators on
Lucassen PJ. Opposite effects of early maternal deprivation on neuro- synaptic plasticity in adult rat hippocampal dentate gyrus. Neurobiol
genesis in male versus female rats. PLoS ONE 2009; 4: e3675. Learn Mem 2009; 92: 292–300.
120 Roceri M, Hendriks W, Racagni G, Ellenbroek BA, Riva MA. Early mater- 138 Smit-Rigter LA, Champagne DL, van Hooft JA. Lifelong impact of varia-
nal deprivation reduces the expression of BDNF and NMDA receptor tions in maternal care on dendritic structure and function of cortical
subunits in rat hippocampus. Mol Psychiatry 2002; 7: 609–616. layer 2/3 pyramidal neurons in rat offspring. PLoS ONE 2009; 4:
121 Kikusui T, Ichikawa S, Mori Y. Maternal deprivation by early weaning e5167.
increases corticosterone and decreases hippocampal BDNF and neuro- 139 Bagot RC, Tse YC, Nguyen HB, Wong AS, Meaney MJ, Wong TP. Mater-
genesis in mice. Psychoneuroendocrinology 2009; 34: 762–772. nal care influences hippocampal N-methyl-D-aspartate receptor func-
122 Oomen CA, Soeters H, Audureau N, Vermunt L, van Hasselt FN, Mand- tion and dynamic regulation by corticosterone in adulthood. Biol
ers EM, Joels M, Krugers H, Lucassen PJ. Early maternal deprivation Psychiatry 2012; 72: 491–498.
affects dentate gyrus structure and emotional learning in adult female 140 Rodenas-Ruano A, Chavez AE, Cossio MJ, Castillo PE, Zukin RS. REST-
rats. Psychopharmacology 2011; 214: 249–260. dependent epigenetic remodeling promotes the developmental switch
123 Brunson KL, Kramar E, Lin B, Chen Y, Colgin LL, Yanagihara TK, Lynch in synaptic NMDA receptors. Nat Neurosci 2012; 15: 1382–1390.
G, Baram TZ. Mechanisms of late-onset cognitive decline after early- 141 Bredy TW, Humpartzoomian RA, Cain DP, Meaney MJ. Partial reversal
life stress. J Neurosci 2005; 25: 9328–9338. of the effect of maternal care on cognitive function through environ-
124 Rice CJ, Sandman CA, Lenjavi MR, Baram TZ. A novel mouse model for mental enrichment. Neuroscience 2003; 118: 571–576.
acute and long- lasting consequences of early life stress. Endocrinol- 142 Champagne DL, de Kloet ER, Joels M. Fundamental aspects of the
ogy 2008; 149: 4892–4900. impact of glucocorticoids on the (immature) brain. Sem Fetal Neonatal
125 Gunn BG, Cunningham L, Cooper MA, Corteen NL, Seifi M, Swinny JD, Med 2009; 14: 136–142.
Lambert JJ, Belelli D. Dysfunctional astrocytic and synaptic regulation 143 van Hasselt FN, Tieskens JM, Trezza V, Krugers HJ, Vanderschuren LJ,
of hypothalamic glutamatergic transmission in a mouse model of Joels M. Within- litter variation in maternal care received by individual
early-life adversity: relevance to neurosteroids and programming of pups correlates with adolescent social play behavior in male rats. Phys-
the stress response. J Neurosci 2013; 33: 19534–19554. iol Behav 2012; 106: 701–706.

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology
722 S. Maccari et al.

144 van Hasselt FN, Boudewijns ZS, van der Knaap NJ, Krugers HJ, Joels M. ylation and transcriptional gene silencing. Development 2012; 139:
Maternal care received by individual pups correlates with adult CA1 2792–2803.
dendritic morphology and synaptic plasticity in a sex-dependent 165 Flanagan JM, Wild L. An epigenetic role for noncoding RNAs and intra-
manner. J Neuroendocrinol 2012; 24: 331–340. genic DNA methylation. Genome Biol 2007; 8: 307.
145 van Hasselt FN, Cornelisse S, Zhang TY, Meaney MJ, Velzing EH, Kru- 166 D’Alessio AC, Szyf M. Epigenetic tete-a-tete: the bilateral relationship
gers HJ, Joels M. Adult hippocampal glucocorticoid receptor expression between chromatin modifications and DNA methylation. Biochem Cell
and dentate synaptic plasticity correlate with maternal care received Biol 2006; 84: 463–476.
by individuals early in life. Hippocampus 2012; 22: 255–266. 167 Lister R, Mukamel EA, Nery JR, Urich M, Puddifoot CA, Johnson ND,
146 van Hasselt FN, de Visser L, Tieskens JM, Cornelisse S, Baars AM, Lavrij- Lucero J, Huang Y, Dwork AJ, Schultz MD, Yu M, Tonti-Filippini J, Heyn
sen M, Krugers HJ, van den Bos R, Joels M. Individual variations in H, Hu S, Wu JC, Rao A, Esteller M, He C, Haghighi FG, Sejnowski TJ,
maternal care early in life correlate with later life decision-making and Behrens MM, Ecker JR. Global epigenomic reconfiguration during mam-
c-fos expression in prefrontal subregions of rats. PLoS ONE 2012; 7: malian brain development. Science 2013; 341: 1237905.
e37820. 168 Gruenbaum Y, Cedar H, Razin A. Substrate and sequence specificity of
147 Bertram C, Khan O, Ohri S, Phillips DI, Matthews SG, Hanson MA. a eukaryotic DNA methylase. Nature 1982; 295: 620–622.
Transgenerational effects of prenatal nutrient restriction on cardiovas- 169 Guo JU, Su Y, Shin JH, Shin J, Li H, Xie B, Zhong C, Hu S, Le T, Fan G,
cular and hypothalamic-pituitary-adrenal function. J Physiology 2008; Zhu H, Chang Q, Gao Y, Ming GL, Song H. Distribution, recognition and
586: 2217–2229. regulation of non-CpG methylation in the adult mammalian brain. Nat
148 Dunn GA, Bale TL. Maternal high-fat diet promotes body length Neurosci 2014; 17: 215–222.
increases and insulin insensitivity in second-generation mice. Endocri- 170 Comb M, Goodman HM. CpG methylation inhibits proenkephalin gene
nology 2009; 150: 4999–5009. expression and binding of the transcription factor AP-2. Nucleic Acids
149 Dunn GA, Bale TL. Maternal high-fat diet effects on third-generation Res 1990; 18: 3975–3982.
female body size via the paternal lineage. Endocrinology 2011; 152: 171 Nan X, Ng HH, Johnson CA, Laherty CD, Turner BM, Eisenman RN, Bird
2228–2236. A. Transcriptional repression by the methyl-CpG-binding protein
150 Drake AJ, Walker BR, Seckl JR. Intergenerational consequences of fetal MeCP2 involves a histone deacetylase complex. Nature 1998; 393:
programming by in utero exposure to glucocorticoids in rats. Am J 386–389.
Physiol Regul Integr Comp Physiol 2005; 288: R34–R38. 172 Ramchandani S, Bhattacharya SK, Cervoni N, Szyf M. DNA methylation
151 Rodgers AB, Morgan CP, Bronson SL, Revello S, Bale TL. Paternal stress is a reversible biological signal. Proc Natl Acad Sci USA 1999; 96:
exposure alters sperm microRNA content and reprograms offspring 6107–6112.
HPA stress axis regulation. J Neurosci 2013; 33: 9003–9012. 173 Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M. A mammalian
152 Dietz DM, Laplant Q, Watts EL, Hodes GE, Russo SJ, Feng J, Oosting RS, protein with specific demethylase activity for mCpG DNA. Nature
Vialou V, Nestler EJ. Paternal transmission of stress-induced patholo- 1999; 397: 579–583.
gies. Biol Psychiatry 2011; 70: 408–414. 174 Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of
153 Dias BG, Ressler KJ. Parental olfactory experience influences behavior Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and
and neural structure in subsequent generations. Nat Neurosci 2014; inner cell mass specification. Nature 2010; 466: 1129–1133.
17: 89–96. 175 Valinluck V, Sowers LC. Endogenous cytosine damage products alter
154 Szyf M. How do environments talk to genes? Nat Neurosci 2013; 16: the site selectivity of human DNA maintenance methyltransferase
2–4. DNMT1. Cancer Res 2007; 67: 946–950.
155 Waddington CH. Canalization of development and genetic assimilation 176 Inoue A, Zhang Y. Replication-dependent loss of 5-hydroxymethylcyto-
of acquired characters. Nature 1959; 183: 1654–1655. sine in mouse preimplantation embryos. Science 2011; 334: 194.
156 Szyf M. The early life social environment and DNA methylation: DNA 177 Razin A, Szyf M, Kafri T, Roll M, Giloh H, Scarpa S, Carotti D, Cantoni
methylation mediating the long-term impact of social environments GL. Replacement of 5-methylcytosine by cytosine: a possible mecha-
early in life. Epigenetics 2011; 6: 971–978. nism for transient DNA demethylation during differentiation. Proc Natl
157 Hotchkiss RD. The quantitative separation of purines, pyrimidines, and Acad Sci USA 1986; 83: 2827–2831.
nucleosides by paper chromatography. J Biol Chem 1948; 175: 178 Jost JP. Nuclear extracts of chicken embryos promote an active deme-
315–332. thylation of DNA by excision repair of 5-methyldeoxycytidine. Proc
158 Kriaucionis S, Heintz N. The nuclear DNA base 5-hydroxymethylcytosine Natl Acad Sci USA 1993; 90: 4684–4688.
is present in Purkinje neurons and the brain. Science 2009; 324: 179 Guo JU, Su Y, Zhong C, Ming GL, Song H. Hydroxylation of 5-methyl-
929–930. cytosine by TET1 promotes active DNA demethylation in the adult
159 Wade PA, Pruss D, Wolffe AP. Histone acetylation: chromatin in action. brain. Cell 2011; 145: 423–434.
Trends Biochem Sci 1997; 22: 128–132. 180 Barreto G, Schafer A, Marhold J, Stach D, Swaminathan SK, Handa V,
160 Jenuwein T. Re-SET-ting heterochromatin by histone methyltransferas- Doderlein G, Maltry N, Wu W, Lyko F, Niehrs C. Gadd45a promotes epi-
es. Trends Cell Biol 2001; 11: 266–273. genetic gene activation by repair-mediated DNA demethylation. Nature
161 Shilatifard A. Chromatin modifications by methylation and ubiquitina- 2007; 445: 671–675.
tion: implications in the regulation of gene expression. Annu Rev 181 Rai K, Huggins IJ, James SR, Karpf AR, Jones DA, Cairns BR. DNA
Biochem 2006; 2432: 243–269. demethylation in zebrafish involves the coupling of a deaminase, a gly-
162 Shiio Y, Eisenman RN. Histone sumoylation is associated with tran- cosylase, and gadd45. Cell 2008; 135: 1201–1212.
scriptional repression. Proc Natl Acad Sci USA 2003; 100: 182 Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ,
13225–13230. Szyf M. Reversal of maternal programming of stress responses in adult
163 Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. offspring through methyl supplementation: altering epigenetic marking
Cell 2004; 116: 281–297. later in life. J Neurosci 2005; 25: 11045–11054.
164 Mohammad F, Pandey GK, Mondal T, Enroth S, Redrup L, Gyllensten U, 183 Weaver IC, D’Alessio AC, Brown SE, Hellstrom IC, Dymov S, Sharma S,
Kanduri C. Long noncoding RNA-mediated maintenance of DNA meth- Szyf M, Meaney MJ. The transcription factor nerve growth fac-

© 2014 British Society for Neuroendocrinology Journal of Neuroendocrinology, 2014, 26, 707–723
Consequences of early-life adversity 723

tor-inducible protein a mediates epigenetic programming: altering changes DNA methylation States in male organ systems: multigenera-
epigenetic marks by immediate-early genes. J Neurosci 2007; 27: tional effects. Endocrinology 2012; 153: 3269–3283.
1756–1768. 194 Crudo A, Suderman M, Moisiadis VG, Petropoulos S, Kostaki A, Hal-
184 McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonte B, Szyf M, lett M, Szyf M, Matthews SG. Glucocorticoid programming of the
Turecki G, Meaney MJ. Epigenetic regulation of the glucocorticoid fetal male hippocampal epigenome. Endocrinology 2013; 154: 1168–
receptor in human brain associates with childhood abuse. Nat Neurosci 1180.
2009; 12: 342–348. 195 Provencal N, Suderman MJ, Guillemin C, Massart R, Ruggiero A, Wang
185 Roth TL, Lubin FD, Funk AJ, Sweatt JD. Lasting epigenetic influence of D, Bennett AJ, Pierre PJ, Friedman DP, Cote SM, Hallett M, Tremblay
early-life adversity on the BDNF gene. Biol Psychiatry 2009; 65: 760– RE, Suomi SJ, Szyf M. The signature of maternal rearing in the methy-
769. lome in rhesus macaque prefrontal cortex and T cells. J Neurosci 2012;
186 Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, Fischer D, 32: 15626–15642.
Holsboer F, Wotjak CT, Almeida OF, Spengler D. Dynamic DNA methyla- 196 Mehta D, Klengel T, Conneely KN, Smith AK, Altmann A, Pace TW, Rex-
tion programs persistent adverse effects of early-life stress. Nat Haffner M, Loeschner A, Gonik M, Mercer KB, Bradley B, Muller-My-
Neurosci 2009; 12: 1559–1566. hsok B, Ressler KJ, Binder EB. Childhood maltreatment is associated
187 Klengel T, Mehta D, Anacker C, Rex-Haffner M, Pruessner JC, Pariante with distinct genomic and epigenetic profiles in posttraumatic stress
CM, Pace TW, Mercer KB, Mayberg HS, Bradley B, Nemeroff CB, Holsb- disorder. Proc Natl Acad Sci USA 2013; 110: 8302–8307.
oer F, Heim CM, Ressler KJ, Rein T, Binder EB. Allele-specific FKBP5 197 Borghol N, Suderman M, McArdle W, Racine A, Hallett M, Pembrey M,
DNA demethylation mediates gene-childhood trauma interactions. Nat Hertzman C, Power C, Szyf M. Associations with early-life socio-eco-
Neurosci 2013; 16: 33–41. nomic position in adult DNA methylation. Int J Epidemiol 2012; 41:
188 Szyf M. The early-life social environment and DNA methylation. Clin 62–74.
Genet 2012; 81: 341–349. 198 Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin AM.
189 McGowan PO, Sasaki A, Huang TC, Unterberger A, Suderman M, Ernst Prenatal exposure to maternal depression, neonatal methylation of
C, Meaney MJ, Turecki G, Szyf M. Promoter-wide hypermethylation of human glucocorticoid receptor gene (NR3C1) and infant cortisol stress
the ribosomal RNA gene promoter in the suicide brain. PLoS ONE responses. Epigenetics 2008; 3: 97–106.
2008; 3: e2085. 199 Perroud N, Paoloni-Giacobino A, Prada P, Olie E, Salzmann A, Nicas-
190 Weaver IC, Meaney MJ, Szyf M. Maternal care effects on the hippo- tro R, Guillaume S, Mouthon D, Stouder C, Dieben K, Huguelet P,
campal transcriptome and anxiety-mediated behaviors in the offspring Courtet P, Malafosse A. Increased methylation of glucocorticoid
that are reversible in adulthood. Proc Natl Acad Sci USA 2006; 103: receptor gene (NR3C1) in adults with a history of childhood mal-
3480–3485. treatment: a link with the severity and type of trauma. Transl Psychi-
191 McGowan PO, Suderman M, Sasaki A, Huang TC, Hallett M, Meaney atry 2011; 1: e59.
MJ, Szyf M. Broad epigenetic signature of maternal care in the brain 200 Tyrka AR, Price LH, Marsit C, Walters OC, Carpenter LL. Childhood
of adult rats. PLoS ONE 2011; 6: e14739. adversity and epigenetic modulation of the leukocyte glucocorticoid
192 Suderman M, McGowan PO, Sasaki A, Huang TC, Hallett MT, Meaney receptor: preliminary findings in healthy adults. PLoS ONE 2012; 7:
MJ, Turecki G, Szyf M. Conserved epigenetic sensitivity to early life e30148.
experience in the rat and human hippocampus. Proc Natl Acad Sci 201 Melas PA, Wei Y, Wong CC, Sjoholm LK, Aberg E, Mill J, Schalling M,
USA 2012; 109(Suppl. 2): 17266–17272. Forsell Y, Lavebratt C. Genetic and epigenetic associations of MAOA
193 Crudo A, Petropoulos S, Moisiadis VG, Iqbal M, Kostaki A, Machnes Z, and NR3C1 with depression and childhood adversities. Int J Neuropsy-
Szyf M, Matthews SG. Prenatal synthetic glucocorticoid treatment chopharmacol 2013; 16: 1513–1528.

Journal of Neuroendocrinology, 2014, 26, 707–723 © 2014 British Society for Neuroendocrinology

You might also like