ArticlePDF AvailableLiterature Review

Neuroendocrinology of childbirth and mother–child attachment: The basis of an etiopathogenic model of perinatal neurobiological disorders

Authors:
  • European Institute of Perinatal Mental Health

Abstract and Figures

This review focuses on the neuroendocrine mechanisms in the mother and the newborn that are involved in the generation and consolidation of mother-child attachment. The role that different hormones and neurotransmitters play on the regulation of these mechanisms during parturition, the immediate postpartum period and lactation is discussed. Interferences in the initiation of mother-child attachment may have potential long-term effects for the behavior and affection of the newborn. Therefore, the possible consequences of alterations in the physiological neuroendocrine mechanisms of attachment, caused by elective Cesarean section, intrapartum hormonal manipulations, preterm delivery, mother-infant postpartum separation and bottle-feeding instead of breastfeeding are also discussed.
Hormonal actions during parturition and early postpartum involved in the regulation of maternal care and the bonding between the mother and her newborn. (A) Vaginal delivery results in increased brain oxytocin release which will facilitate mother-infant bonding. In addition, oxytocin during labor changes the effect of GABA in the fetal brain from being excitatory to be inhibitory, allowing a protective effect of GABA from delivery hypoxia. In the baby, the passage of the head through the birth canal is accompanied by a massive release of catecholamines and vasopressin that probably facilitate newborn physiological adaptation after delivery. Furthermore, maternal uterine contractions during labor facilitate neonatal olfactory learning in the newborn. (B) In the early postpartum period, noradrenaline, oxytocin, vasopressin and prolactin promote olfactory learning in the mother and maternal care. In rodents corticosterone affects neural plasticity in different brain regions, decreases hippocampal neurogenesis and impairs spatial learning. In humans, higher cortisol levels in the mother are associated with higher preference for her newborn body odor. Skin-to-skin contact of the mother with her newborn and the initiation of breastfeeding promote increased levels of oxytocin in the mother and the baby. Oxytocin contributes to mother-infant affect synchrony, promoting the attachment of the mother and the newborn. This attachment is essential for the regulation of emotions, stress reactivity, metabolic adaptation and social and cognitive development of the baby.
… 
Content may be subject to copyright.
Review
Neuroendocrinology of childbirth and mother–child attachment: The
basis of an etiopathogenic model of perinatal neurobiological disorders
Ibone Olza-Fernández
a
, Miguel Angel Marín Gabriel
b
, Alfonso Gil-Sanchez
c
, Luis M. Garcia-Segura
d,
,
Maria Angeles Arevalo
d
a
Department of Psychiatry, Autonomous University of Madrid, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
b
Department of Pediatrics, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
c
Unidad Docente de Salud Mental de la Región de Murcia, Hospital General Universitario Santa María del Rosell de Cartagena, Murcia, Spain
d
Instituto Cajal, CSIC, Madrid, Spain
article info
Article history:
Available online 3 April 2014
Keywords:
Autism
Cesarean section
Childbirth
Maternal behavior
Mother–child attachment
Noradrenaline
Oxytocin
Progesterone
Prolactin
Vasopressin
abstract
This review focuses on the neuroendocrine mechanisms in the mother and the newborn that are involved
in the generation and consolidation of mother–child attachment. The role that different hormones and
neurotransmitters play on the regulation of these mechanisms during parturition, the immediate post-
partum period and lactation is discussed. Interferences in the initiation of mother–child attachment
may have potential long-term effects for the behavior and affection of the newborn. Therefore, the pos-
sible consequences of alterations in the physiological neuroendocrine mechanisms of attachment, caused
by elective Cesarean section, intrapartum hormonal manipulations, preterm delivery, mother–infant
postpartum separation and bottle-feeding instead of breastfeeding are also discussed.
Ó2014 Elsevier Inc. All rights reserved.
1. Introduction: Childbirth as a neurobiological and
neuroendocrine event
Physiological changes in the dynamics and mechanics of labor
in women have been thoroughly studied (Liao et al., 2005). How-
ever, the peripartal neurohormonal scenery in the brain of the
mother, the fetus and the newborn has received little attention.
Here we will approach human childbirth as a neurohormonal event
rather than a mechanical one and we will specifically focus on the
peripartal neuroendocrine mechanisms that participate in the gen-
eration of mother–child attachment. We will also consider possible
disrupting effects that some obstetrical interventions during the
peripartal period may have on the neuroendocrine mechanisms
of mother–child attachment, as well as its possible long-term con-
sequences for the newborn. Birth outcomes have traditionally been
measured in terms of maternal and neonatal morbidity in the short
term, indicators that may not be valid to study the impact of hypo-
thetical changes in the brain during childbirth or the early postna-
tal period. This impact may only be manifested later in life, during
adolescence or even in adulthood. Understanding the neuroendo-
crine events involved in mother–child attachment that happen in
both maternal and fetal brains during the peripartum period may
allow the building of an etiopathogenic model of some mental
disorders that would be better understood as the consequence of
peripartal neuroendocrine alterations.
Motherhood entails neurochemical, morphological and func-
tional brain changes designed to ensure the survival of newborns.
Hormonal changes during different life stages are associated with
the regulation of specific neuroplastic events that generate appro-
priate physiological and behavioral responses in adaptation to and
in prediction of changing life conditions (Garcia-Segura, 2009).
Pregnancy is one of the life stages characterized by prominent
hormonal changes and is also a period of enhanced-neuroplastic
changes that result even in the reduction of brain size in humans
(Oatridge et al., 2002) and of hippocampal volume in rats (Galea
et al., 2000). The neuroendocrine and neuroplastic events during
pregnancy that facilitate the onset of adequate maternal behaviors
have been extensively studied and characterized in different mam-
malian species. These events, which involve different actions of
steroid hormones, neurosteroids, prolactin, oxytocin, vasopressin,
catecholamines and endorphins (Keverne, 1988; Torner and
Neumann, 2002; Russell et al., 2003; Douglas, 2005; Brunton
http://dx.doi.org/10.1016/j.yfrne.2014.03.007
0091-3022/Ó2014 Elsevier Inc. All rights reserved.
Corresponding author. Address: Instituto Cajal, CSIC, Avenida Doctor Arce 37,
E-28002 Madrid, Spain. Fax: +34 915854754.
E-mail address: lmgs@cajal.csic.es (L.M. Garcia-Segura).
Frontiers in Neuroendocrinology 35 (2014) 459–472
Contents lists available at ScienceDirect
Frontiers in Neuroendocrinology
journal homepage: www.elsevier.com/locate/yfrne
et al., 2008; Brunton and Russell, 2008, 2010), will not be reviewed
in the present paper.
In this review we will specifically focus on the hormonal cas-
cade in the maternal and the newborn brain, at parturition, the
early postpartum period and lactation, which primes the mother
and the newborn for attachment initiation and consolidation
(Fig. 1). Attachment theory, proposed by Bowlby, has become the
dominant model of human emotional and social development.
According to Bowlby, attachment between the infant and his or
her mother is an innate biological response that increases the
probability of survival (Bowlby, 1978). Our hypothesis is that in
humans, like in other mammalian species (Mogi et al., 2011),
peripartal neuroendocrine events play an essential role in the ini-
tiation of the bonding of the mother and the newborn immediately
after birth. A disruption in these neuroendocrine events, even
when intervention is necessary due to serious medical reasons, is
likely to have not only a short-term impact on mother–child
attachment but also long-term effects in the newborn, increasing
the risk of behavioral alterations or mental health problems that
may have not yet been causally related to a peripartal origin. Thus,
we will also review in the present paper several potential etiopath-
ogenic neurohormonal scenarios during labor and the early
postpartum period that are associated with neuroendocrine modi-
fications, such as elective Cesarean section, intrapartum hormonal
manipulations, preterm delivery, mother–infant postpartum sepa-
ration and bottle-feeding instead of breastfeeding (Fig. 2).
2. Neuroendocrine events at parturition that affect
mother–child attachment
2.1. Neuroendocrine events in the mother at parturition
2.1.1. Oxytocin
Studies in rodents and sheep indicate that the stimulation of the
vagina and cervix at birth plays an important role in the induction
of maternal behavior (Keverne et al., 1983; Yeo and Keverne, 1986;
Kendrick and Keverne, 1991). Vaginal and cervix stimulation also
triggers in sheep the formation of olfactory recognition memory
that is essential for the selective recognition of lambs and for the
formation of maternal bonding (Keverne et al., 1983; Kendrick
et al., 1991b). These effects on maternal behavior and olfactory rec-
ognition may be mediated by the increased levels of oxytocin in
the brain caused by the stimulation of the vagina and cervix. In-
deed, one of the most important neuroendocrine events in the
mother at parturition in relation with mother–child attachment
is the activation of the oxytocinergic system. In rodents, gonadal
steroids prime the oxytocin system to become activated in prepa-
ration of birth and maternal behavior (Crowley et al., 1995; Amico
et al., 1997; Windle et al., 2006; Donner et al., 2007). However,
oxytocinergic neurons remain at rest during pregnancy, avoiding
premature birth (Brunton and Russell, 2010). Several mechanisms
and hormonal systems contribute to maintain oxytocinergic neu-
rons at rest until parturition (Brunton et al., 2014). These include
actions of central opioids, which contribute to restrain premature
oxytocin release by acting on
l
-receptors on magnocellular oxyto-
cinergic neurons or on presynaptic inputs to these neurons (Dondi
et al., 1991; Douglas et al., 1993, 1995; Wigger et al., 1999). In
addition, the progesterone metabolite allopregnanolone, prevents
the release of oxytocin in late gestation through potentiation of
the inhibitory effect of GABA
A
receptors on magnocellular oxyto-
cinergic neurons (Brunton and Russell, 2010; Brunton et al.,
2014) and by inducing opioid inhibition over these neurons
(Brunton et al., 2012, 2014).
While the activation of the oxytocinergic system is inhibited
during pregnancy by the above mentioned mechanisms, its activa-
tion is necessary for parturition and for the onset of lactation and
maternal behavior. Consistently with the essential role of oxytocin
release by magnocellular neurons for parturition and lactation,
functional and anatomical plastic changes in oxytocin neurons
and associated glial cells are manifested around parturition and
the onset of lactation in rodents (Leng et al., 1999; Theodosis and
Poulain, 2001; Tasker et al., 2002; Koksma et al., 2005; Theodosis
et al., 2006). These neuroplastic events include changes in oxytocin
immunoreactivity (Jirikowski et al., 1989), in neuronal firing
(Lincoln and Wakerley, 1974; Leng et al., 1999), in the number of
glutamatergic and GABAergic synaptic inputs (Theodosis and
Poulain, 2001), in the release of glutamate and GABA (Stern et al.,
Parturition
Catecholamines
Vasopressin
Olfactory
learning
Oxytocin
Prolactin
Endorphins
Facilitation of
mother-infant attachment
Vasopressin
Prolactin
Oxytocin
Noradrenaline
Odor information
Maternal care
Cortisol
Neurogenesis
Spatial learning
-Emotions
-Stress reactivity
-Metabolic adaptation
-Social and cognitive
development
Oxytocin
Early Postpartum
Attachment
A
B
Fig. 1. Hormonal actions during parturition and early postpartum involved in the
regulation of maternal care and the bonding between the mother and her newborn.
(A) Vaginal delivery results in increased brain oxytocin release which will facilitate
mother–infant bonding. In addition, oxytocin during labor changes the effect of
GABA in the fetal brain from being excitatory to be inhibitory, allowing a protective
effect of GABA from delivery hypoxia. In the baby, the passage of the head through
the birth canal is accompanied by a massive release of catecholamines and
vasopressin that probably facilitate newborn physiological adaptation after deliv-
ery. Furthermore, maternal uterine contractions during labor facilitate neonatal
olfactory learning in the newborn. (B) In the early postpartum period, noradren-
aline, oxytocin, vasopressin and prolactin promote olfactory learning in the mother
and maternal care. In rodents corticosterone affects neural plasticity in different
brain regions, decreases hippocampal neurogenesis and impairs spatial learning. In
humans, higher cortisol levels in the mother are associated with higher preference
for her newborn body odor. Skin-to-skin contact of the mother with her newborn
and the initiation of breastfeeding promote increased levels of oxytocin in the
mother and the baby. Oxytocin contributes to mother–infant affect synchrony,
promoting the attachment of the mother and the newborn. This attachment is
essential for the regulation of emotions, stress reactivity, metabolic adaptation and
social and cognitive development of the baby.
460 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
2000; Tasker et al., 2002) and in the expression, function and
clustering of postsynaptic receptors (Brussaard et al., 1997, 1999;
Koksma et al., 2005). Synaptic changes in oxytocin neurons are
associated with the remodeling of astroglia processes. Glial pro-
cesses, which normally separate the oxytocin-containing magno-
cellular neurons, are retracted during pregnancy, birth, and
lactation, allowing electrical coupling and then the pulsatile re-
lease of oxytocin (Theodosis and Poulain, 2001; Carter, 2014).
In women, the best known effect of peripheral oxytocin during
childbirth is to increase uterine contractions with a positive feed-
back, which will culminate with the ejection reflex, a physiological
process that makes childbirth successful mechanically and explain
the rapid deliveries (Odent, 2009). The importance of spontaneous
birth reflex explains the detrimental effects of sustained Valsalva
bearing down efforts and supine positioning (Roberts and Hanson,
2007). It also explains why directed pushing during childbirth is
not associated with optimal results (Brancato et al., 2008; Hanson,
2009).
In addition, vaginal delivery results in increased levels of oxyto-
cin not only in the periphery but also in the brain, as indicated by
animal studies showing increased levels of the hormone at partu-
rition in the cerebrospinal fluid and the paraventricular nucleus
of sheep (Kendrick et al., 1991a; Da Costa et al., 1996) and in the
brain of rats (Neumann et al., 1991, 1993a,b; Landgraf et al.,
1992). In addition, oxytocin receptor expression in rats is also in-
creased at parturition in different brain regions, such as the bed
nucleus of the stria terminalis and medial preoptic area (Caughey
et al., 2011). In humans there is also a significant increase in oxy-
tocin levels in the cerebrospinal fluid during normal labor (Takeda
et al., 1985). Interestingly, changes in the central and peripheral
levels of oxytocin during parturition in women do not show the
same pattern. Thus, while plasma levels of oxytocin increase before
labor, oxytocin levels in the cerebrospinal fluid are higher after la-
bor than before labor. This suggests that plasma and brain levels of
oxytocin are regulated by different mechanisms and probably have
different functions (Takeda et al., 1985).
The central increase in oxytocin levels during delivery in
women may affect maternal behavior, since it is well known that
central actions of oxytocin are necessary for the expression of
an adequate maternal behavior, at least in sheep and rodents
(Kendrick et al., 1997;Da Costa et al., 1996; Bosch and Neumann,
2012). In particular, the central increase in oxytocin levels may
contribute to the generation of mother–infant bonding, which
may be associated with an increase activation of oxytocin and
dopamine pathways in the brain (Nissen et al., 1998; Strathearn,
2011). High levels of endogenous oxytocin in humans are corre-
lated with feelings of well-being, confidence, love, social memory
and even mystical experiences (Baumgartner et al., 2008;
Heinrichs et al., 2009; Keri and Kiss, 2010; Ishak et al., 2011).
Feelings of love, transpersonal and transcendental experiences
during labor are often narrated by mothers from different cultures
(Callister et al., 1999; Callister, 2004). These psychological changes
may facilitate the mother–infant bonding process.
2.1.2. Cortisol
Another hormone that changes during parturition in women
and that may affect the interaction of the mother with her baby
is cortisol. In women, the levels of cortisol are increased during la-
bor (Willcox et al., 1985; Campbell et al., 1987; Ohana et al., 1996).
This increase in cortisol levels may also contribute to the develop-
ment of mother–child attachment, since circulating or salivary cor-
tisol levels in human mothers have been positively correlated with
several aspects of maternal behavior (Fleming et al., 1987, 1997;
Stallings et al., 2001; Krpan et al., 2005), including the recognition
and attraction for baby odors (Fleming et al., 1997). Corticosterone
also increases maternal behavior in postpartum rats (Graham et al.,
2006) (see Section 3.1).
Parturition Early Postpartum
Cesarean section
Neurohormonal manipulations
Anesthetic drugs (opioid agonists)
Mother-infant
separation
Synthetic oxytocin
Oxytocin receptor antagonists
Preterm delivery
Bottle-feeding
Peripartal neurohormonal disruptions
Maternal Psychopathology:
PPD, PTSD.
Maternal Care Hippocampal development
Developmental Psychopathology:
Attachment , autism, ADHD, anxiety,
feeding, learning disabilities.
Attachment disruption
Fig. 2. Altered neurohormonal scenarios that may affect mother–infant attachment, according to the chronological time when hormonal alterations happen during
childbirth. Modifications in the neuroendocrine regulation at perinatal period are likely to increase susceptibility to problems in the newborn latter in life. Several
etiopatothogenic scenarios during labor and the postpartum period can be envisaged. These include interventions during parturition, such as Cesarean section, administration
of synthetic oxytocin, oxytocin receptor antagonists or opioids, preterm delivery, mother–infant separation after delivery or the substitution of breastfeeding by bottle-
feeding. All these conditions and circumstances result in modifications in the physiological levels of some key hormones, such as oxytocin, which are involved in the process
of mother–infant attachment. These peripartal neurohormonal disruptions may contribute to alterations in maternal care and be a risk factor for the development of affective
disorders in the mother, such as postpartum depression (PPD) or posttraumatic stress disorder (PTSD). Hormonal alterations and the disruption of attachment may also affect
the development of some regions in the brain in the newborn, such as the hippocampus and may represent a risk factor for the development of autism, attention deficit
hyperactive disorder (ADHD), anxiety, feeding disorders, learning disabilities or other mental disorders.
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 461
2.2. Neuroendocrine events in the fetus at parturition
An investigation of the mechanism of labor using real time MRI
confirmed that human fetuses must negotiate a curve to be born. An
MRI video from that study shows clearly the strong physical com-
pression that the fetal brain undergoes during labor (Bamberg
et al., 2012). This intrapartum mechanical compression of the fetal
brain probably leads to a massive neurohormonal release and to
electrophysiological changes that have not yet been thoroughly
studied. In rats, a dramatic surge of neurotransmitters, neuroster-
oids, neuropeptides, and neuromodulators occurs during the birth
process and shortly thereafter, facilitating physiological processes
such as respiration (Ronca and Alberts, 1995) and suckling (Abel
et al., 1998). In humans, the passage of the baby head through the
birth canal is accompanied by a substantial activation of the sympa-
thoadrenal axis and the enhanced release of noradrenaline, cortisol
and vasopressin (Gennser et al., 1977; Puolakka et al., 1983; Costa
et al., 1988; Wellmann et al., 2010). This situation has been called
by some authors ‘‘the stress of labor’’, which is both beneficial
and necessary for newborns (Lagercrantz and Slotkin, 1986). It
has been hypothesized that this eustressor acts to prime the fetal
genome to trigger optimal responses to extra uterine life (Dahlen
et al., 2013). The massive sympathoadrenal activation stimulates
lung fluid reabsorption and facilitates newborn physiological
adaptation after delivery. It promotes lung maturity, increases
blood flow, regulates circulating cytokine levels, mobilizes fuel
and activates the central nervous system (Chen et al., 1998;
Malamitsi-Puchner et al., 2005; Miller et al., 2005; Yektaei-Karin
et al., 2007). The alterations produced by vaginal birth are resolved
after birth by vagal stimulation produced by skin to skin contact and
early suckling (Bystrova et al., 2003; Ferber and Makhoul, 2004).
The increase in noradrenaline levels in the newborn at parturi-
tion (Puolakka et al., 1983; Costa et al., 1988) may facilitate
mother–child attachment, since noradrenaline levels in the new-
born are positively correlated with olfactory learning shortly after
birth (Varendi et al., 2002). During a natural childbirth the odor of
the mother is supposed to be the first biologically relevant odor
that the newborn confronts. Therefore, the increase in noradrena-
line levels caused by vaginal delivery may contribute to the identi-
fication of maternal odor and to the consequent establishment of
the attachment of the newborns with their mothers. This is sug-
gested by the findings of Varendi et al. (2002), who exposed babies
born by Cesarean section to an odor for 30 min shortly after birth.
Babies from births with uterine labor contractions before Cesarean
delivery showed increased preference for the exposed odor when
exposed to a new odor, compared to babies from births without
uterine contractions. These findings suggest that labor-induced
noradrenaline release facilitates olfactory learning in the newborns
shortly after birth and therefore may facilitate mother–child
attachment.
Vaginal birth is also associated with an extremely steep rise of
circulating vasopressin in the newborn (Schubert et al., 1981),
higher than that observed in critically ill adult patients with shock
or brain injury (Wellmann et al., 2010). The consequences of this
massive increase in vasopressin in human newborns have not been
studied, although it has been proposed that the vasopressin rise
may be involved in the analgesic effects caused by vaginal delivery
in human newborns (Wellmann and Buhrer, 2012).
3. Immediate postpartum: Sensitive period
3.1. Mother–child synchrony: Importance of skin-to-skin contact
Humans are born biologically prepared to establish coordinated
interactions from the first hours of life. Right after delivery occurs
the so-called sensitive period, a quiet alertness state that lasts for
about two hours (Bystrova et al., 2009). The neonatal sensitive per-
iod includes the spontaneous onset of breastfeeding in the first two
hours of life. The first hours after birth are also a critical period for
the development of attachment behavior (Mehler et al., 2011).
In humans, randomized controlled studies suggest that the
body of the mother is the natural habitat for the naked infant
(Winberg, 2005). Skin-to-skin contact immediately after delivery
helps the baby to conserve energy, adjust acid–base balance and
breathing and has a calming effect (Uvnäs-Moberg, 1996; Winberg,
2005). It also increases maternal attention to her baby (Winberg,
2005) and reduces cortisol levels in the mother (Handlin et al.,
2009). Skin-to-skin contact with both mothers and fathers reduce
infants’ crying (Christensson et al., 1995; Erlandsson et al., 2007)
and promotes vocal communication between parents and
newborns (Velandia et al., 2010). It is likely that oxytocin released
during the skin-to-skin contact also increases the parental respon-
siveness to infant cues (Velandia et al., 2010). As a consequence of
early skin-to-skin contact, infant regulation of emotions, stress
reactivity, metabolic adaptation, social and cognitive development
and future interaction between mother and infant are promoted
(Bystrova et al., 2009; Velandia et al., 2010). It is the beginning of
the continuous development of mother–child synchrony (Winberg,
2005), facilitated mostly by lactation and oxytocin. Indeed, salivary
oxytocin levels between mothers and infants are significantly cor-
related, being high oxytocin levels associated with greater degree
of mother–infant affect synchrony (Feldman et al., 2010a).
Increased oxytocin levels in the newborn caused by skin-to-skin
contact probably play an important role in the initiation of breast-
feeding. The smell of the breast helps the baby to initiate breast-
feeding. Natural maternal breast odors reduce crying (Doucet
et al., 2007) and elicit approach behavior in newborns whereas
unpleasant odors induce avoidant behavior (Varendi and Porter,
2001). Newborns guided by smell spontaneously seek the nipple
and usually initiate breastfeeding in the first hour of life (Porter
and Winberg, 1999; Varendi et al., 2002) if they are placed in
skin-to-skin contact with their mothers (Widstrom et al., 1987).
In rats, maternal skin-to-skin contact increases central oxytocin
in the pups, facilitating the induction of preference for maternal
odor and the establishment of social affiliation (Kojima and
Alberts, 2011; Kojima et al., 2012). Furthermore, higher levels of
noradrenaline immediately after birth may support better odor
learning in rat pups (Miller and Spear, 2008). Thus, odor exposure
after birth likely occurs under a different internal neurochemical
milieu for the pup odor exposure later in life (Ronca et al., 2006).
Indeed, the neonatal surge of noradrenaline may induce an olfac-
tory imprinting-like occurrence in mammals without the need
for an explicit reinforcer to form a lasting odor preference (Sullivan
et al., 1991). This is also probably the case in humans, in which nor-
adrenaline released in the newborn during labor facilitates learn-
ing and recognition of the mother odor (Varendi et al., 2002).
In most mammals, pup odors elicit maternal behavior in the
mother (Kinsley and Amory-Meyer, 2011; Levy et al., 2004).
Human mothers also show changes in olfaction and recognize
the odors of their babies as pleasant. Interestingly, human mothers
have increased oxytocin levels during the first hour after delivery
(Nissen et al., 1995), corresponding to the sensitive period. Oxyto-
cin plays a key role in the processing of olfactory information by
mothers and the infusion of oxytocin in the olfactory bulb
produces a rapid onset of maternal behavior in rats. This, in turn,
modulates the release of noradrenaline, which is involved in olfac-
tory learning not only in the newborn but also in the mother (Levy
et al., 2004; Kinsley and Amory-Meyer, 2011). In rats corticoste-
rone enhances maternal behavior and maternal memory (Graham
et al., 2006). In women, cortisol plays an important role in the
attraction to the body odor of their infants in human mothers.
462 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
Indeed, postpartum cortisol levels in women are positively corre-
lated with attraction for the odor of their own babies and with
the ability to recognize their own baby’s odor (Fleming et al.,
1997).
Vasopressin is also involved in maternal behavior, regulating
maternal care and maternal aggression (Bosch and Neumann,
2008). In rats, the vasopressin system in the mother is activated
around parturition and in lactation, with increased expression of
vasopressin in the paraventricular nucleus of the hypothalamus
and increased release in different structures, such as the bed nu-
cleus of the stria terminalis and the medial preoptic area (Landgraf
et al., 1991; Bosch and Neumann, 2008; Bosch, 2011). This in-
creased release of vasopressin is necessary for the maintenance
of maternal care (Bosch, 2011). In addition, vasopressin release in
the central amygdala correlates with the amount of maternal
aggression, which may be in part mediated by the regulation of
maternal anxiety (Bosch, 2011). In humans, mothers who are car-
riers of RS3 long alleles in the promoter region of the arginine vaso-
pressin receptor 1A gene show decreased maternal sensitivity to
child’s needs and signals and less supportive and guiding behavior
(Avinun et al., 2012; Bisceglia et al., 2012).
Animal studies suggest that the hormonal regulation of mater-
nal behavior may be in part mediated by hormonal regulation of
brain plasticity at parturition and the early postpartum period. In
addition to the plastic changes in the mother hypothalamus asso-
ciated with oxytocinergic and vasopressinergic neurons, there are
other neuroplastic changes during the postpartum period in rats
that may be driven by the decrease in gonadal hormones and the
increase in corticosterone levels (Darnaudery et al., 2007; Leuner
et al., 2007; Green and Galea, 2008; Pawluski et al., 2009, 2011,
2012; Workman et al., 2012). Decreased neurogenesis in the den-
tate gyrus of the dam hippocampus has been observed during
the early postpartum period (Darnaudery et al., 2007; Leuner
et al., 2007; Pawluski and Galea, 2007) in association with an
impairment of spatial learning (Darnaudery et al., 2007). Adrenal-
ectomy and low dose corticosterone administration prevents this
decrease in dentate gyrus neurogenesis in postpartum dams
(Leuner et al., 2007). In contrast, estradiol withdrawal decreases
dentate gyrus neurogenesis after a hormone-simulated pregnancy
(Green and Galea, 2008). Neuroplastic changes during the postpar-
tum period have been also reported in other brain regions (Kinsley
and Lambert, 2008; Kinsley and Amory-Meyer, 2011). For instance,
the cell volume of medial preoptic area neurons is increased after
pregnancy in rats (Keyser-Marcus et al., 2001). In addition, not only
neurons but also glial cells are affected by motherhood. Thus, the
number of oligodendrocytes in the corpus callosum is higher in lac-
tating female rats than in virgin rats (Maheu et al., 2009). Mother-
hood also affects glutamate metabolism in astrocytes in the
cingulate cortex of rats (Salmaso et al., 2011).
Neuroplastic changes during the postpartum period in the
mother brain are influenced by the interaction with their pups
(Rasia-Filho et al., 2004; Kinsley et al., 2006; Pawluski and Galea,
2006; Akbari et al., 2007; Leuner et al., 2007; Kinsley and Lambert,
2008; Furuta and Bridges, 2009; Brus et al., 2010). Thus, maternal
behavior increases neurogenesis in the subventricular zone (Furuta
and Bridges, 2009) and removal of nursing pups prevents the de-
crease in neurogenesis in the dam dentate gyrus (Leuner et al.,
2007). Maternal experience in rats also affects the complexity of
the dendritic arbor and/or density of dendritic spines in CA3 and
CA1 pyramidal neurons in the hippocampus (Kinsley et al., 2006;
Pawluski and Galea, 2006) and in the neurons of the caudate nu-
cleus, the nucleus accumbens shell, the medial preoptic area and
the medial amygdala (Rasia-Filho et al., 2004). Therefore, the inter-
action of the mother with her pups seems to be necessary for the
generation of specific neuroplastic changes in the mother brain
during the postpartum period. It is unknown if similar neuroplastic
changes occur in the brain of human mothers, although the limited
fMRI studies available suggest the existence of functional modifica-
tions in the hypothalamus, amygdala and cerebral cortex in the
brain of women during the postpartum period (Kim et al., 2010;
Lord et al., 2012; Rupp et al., 2014). In addition, these functional
modifications in the brain of postpartum women are influenced
by the interaction with their infants (Kim et al., 2010; Musser
et al., 2012).
3.2. Lactation
In most mammals, the stimulation of the teats by her pups
elicits a modification in the activity of oxytocin magnocellular neu-
rons, resulting in a pulsatile release of oxytocin in the neurohy-
pophysis that leads to periodic milk ejections (Poulain and
Wakerley, 1982). In humans, nipple stimulation also increases oxy-
tocin levels in breastfeeding mothers (Dawood et al., 1981; Amico
and Finley, 1986; Uvnäs-Moberg et al., 1990; Uvnäs-Moberg, 1996;
Matthiesen et al., 2001; White-Traut et al., 2009). In parallel to the
release of oxytocin in the neurohypophysis, there is also an in-
creased intracerebral release of oxytocin during lactation, together
with an increased expression of oxytocin receptors, in specific
brain regions (Kendrick et al., 1988; Moos et al., 1989; Neumann
et al., 1993a,b; Bealer and Crowley, 2001; Veenema and Neumann,
2008). This increase in intracerebral oxytocinergic signaling may
mediate the association between breastfeeding and mother–child
attachment, since central oxytocin promotes maternal care in ro-
dents (Bosch and Neumann, 2012). Although the relation of periph-
eral and central oxytocin is not well understood (Neumann et al.,
2013), in women there is a positive correlation between salivary
oxytocin levels and affectionate maternal behavior during
mother–child interactions (Feldman et al., 2010b). Furthermore,
the interaction with their infants causes higher circulating oxyto-
cin levels and increased activation of mesocorticolimbic reward re-
gions in women with secure attachment than in women with
insecure/dismissing attachment (Strathearn et al., 2009a; Strath-
earn, 2011). Higher circulating oxytocin in fathers is also correlated
with more affectionate and stimulatory behaviors when interact-
ing with their children (Feldman et al., 2010a,b; Gordon et al.,
2010).
Breastfeeding has additional affective consequences for moth-
ers that may also facilitate their interactions with their infants.
Lactation in humans and other mammals reduces physiological
reactivity to various stressors (Stern et al., 1973; Neumann et al.,
1998; Johnstone et al., 2000; Walker et al., 2001; Heinrichs et al.,
2001, 2002; Slattery and Neumann, 2008; Bosch and Neumann,
2012). In rats, lactation reduces corticotropin-releasing factor
mRNA expression in the parvocellular paraventricular hypotha-
lamic neurons (Johnstone et al., 2000; Walker et al., 2001; Klampfl
et al., 2013) and reduced expression of corticotropin-releasing fac-
tor receptors in the pituitary corticotrophs (Neumann et al., 1998).
In women, breastfeeding decreases adrenocorticotropic hormone,
total plasma cortisol and salivary free cortisol and the HPA axis re-
sponse to physical and psychosocial stress (Altemus et al., 1995;
Heinrichs et al., 2001, 2002). Nursing mothers often describe a
state of decreased anxiety (Carter and Altemus, 1997) and interact
more positively with their babies, touching and directing more
smiles towards them, than those using artificial feeding (Duna
and Richards, 1977). Similarly, there is a reduction of the auto-
nomic responses, such as modifications in heart rate and skin con-
ductance, to the cries of babies in lactating versus non-lactating
mothers (Wiesenfeld et al., 1985). This reduction in stress response
associated with breastfeeding, could be seen as an adaptive re-
sponse that protects mothers from an exaggerated response to
stressful stimuli ensuring successful breastfeeding (Carter and
Altemus, 1997).
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 463
It is also important to consider that breastfeeding may have
long-term consequences for the infants. Some studies suggest that
early exclusive breastfeeding may be associated with better cogni-
tive skills latter in life (Isaacs et al., 2010; Hallowell and Spatz,
2012; Deoni et al., 2013; Kafouri et al., 2013). In a study using
MRI, infant breastfeeding was associated with improved develop-
mental growth in late maturing white matter association regions
involved in higher-order cognition, such as executive functioning,
planning, social-emotional functioning and language, domains in
which breastfed infants were also found to have improved perfor-
mance (Deoni et al., 2013; Julvez et al., 2014). In the same study
breastfeeding duration beyond 15 months was associated with im-
proved white matter structure and cognitive performance (Deoni
et al., 2013). In this regard it is important to consider that the dura-
tion of breastfeeding in humans is estimated to range physiologi-
cally from two and a half to seven years of life (Detwyler, 1995)
and that the World Health Organization (WHO) recommends
breastfeeding continuation for up to two years of age or beyond
(WHO, 2003). Breastfeeding statistics indicate that most women
breast their baby soon after birth (95% in Norway, 92% in Australia,
89% in Canada and 76.5% in the United States). In the United States,
of infants born in 2010, 49% were breastfeeding at 6 months and
27% at 12 months (Gionet, 2013; National Center for Chronic
Disease Prevention and Health Promotion, 2013).
4. Potential etiopathogenic neurohormonal scenarios during
labor and the early postpartum period
4.1. The intrapartum period
4.1.1. Omission of all the stages of labor: elective Cesarean section
4.1.1.1. Consequences for the newborn. Several circumstances may
potentially disrupt the neuroendocrine mechanisms of mother–
child attachment during the peripartal period in humans (Fig. 2).
One of these is the elimination of labor in planed Cesarean sections.
The use of Cesarean section for delivery is continuously rising
worldwide, often for non medical reasons. Cesarean rates were of
32.8% in the US in 2011 (Hamilton et al., 2013), 48% in China in
2010 (Lumbiganon et al., 2010; Souza et al., 2010) and 45.9% in
Brazil and 41.9% in Iran in 2008 (Gibbons et al., 2012). In some cul-
tural groups Cesarean can be perceived as a preferred way to deli-
ver (Nuttall, 2000) or even as a safer option, without a medical
reason for it. Although there is a growing concern about the effects
of Cesarean section on brain maturation of infants (Kapellou,
2011), the potential long term consequences of elective Cesarean
sections in both the maternal and the newborn brain have received
little attention. The studies that assess the effects of Cesareans typ-
ically consider only short term health outcomes and physical mor-
bidity (Chiossi et al., 2013).
In planned Cesarean sections the neurohormonal transition is
quite sharp, very different to the neuroendocrine cascade that hap-
pens in a physiological vaginal delivery. As mentioned before, with
vaginal delivery there is a dramatic surge of circulating vasopressin
(approximately 100-fold) in the newborn that does not occur after
programmed Cesarean section (Wellmann et al., 2010; Wellmann
and Buhrer, 2012). The consequences that for the neonate born
by Cesarean section may have the abolition of this increase in vaso-
pressin levels are unknown (see Section 2.2). In addition, catechol-
amines and cortisol levels are relatively low in babies born by
Cesarean section compared to babies born by vaginal delivery
(Gitau et al., 2001; Mears et al., 2004; Miller et al., 2005). These dif-
ferences in catecholamines and cortisol levels may be related with
the reported differences in respiratory distress, a complication that
is more frequent after elective Cesarean sections than after normal
vaginal deliveries (Wankaew et al., 2013). Cesarean section
delivery may also alter gut colonization and immunological devel-
opment (Malamitsi-Puchner et al., 2005; Vogl et al., 2006) and may
cause epigenetic changes in the neonate (Dahlen et al., 2013). In-
deed, an increased DNA-methylation in leucocytes has been de-
tected in infants delivered by Cesarean section compared to
infants vaginally born (Schlinzig et al., 2009), although another
study did not detect an effect of the type of delivery on global
methylation of DNA from blood isolated from umbilical venous
cord at birth (Virani et al., 2012).
A crucial event that occurs in the newborn brain at parturition
is the shift of GABA neurotransmission from being excitatory be-
fore birth to be inhibitory after birth (Cherubini et al., 1991). This
is because during embryonic development the intracellular chlo-
ride concentration [Cl
]
i
in neurons is higher than in postnatal
neurons. Thus, when GABA opens the GABA
A
channel, there is a
chloride efflux and depolarization in prenatal neurons and a chlo-
ride influx and hyperpolarization in postnatal neurons (Cherubini
et al., 1991). Oxytocin plays an essential role in the reduction of
[Cl
]
i
in neurons during parturition (Tyzio et al., 2006; Khazipov
et al., 2008). This action of oxytocin protects the newborn brain
from delivery hypoxia and also exerts analgesic actions in the new-
born (Tyzio et al., 2006; Mazzuca et al., 2011). In addition, the ac-
tion of oxytocin on GABAergic neurons during delivery may also
decrease the risk of long-term deleterious consequences on an en-
hanced neuronal activity and may contribute to prevent the devel-
opment of autism (Tyzio et al., 2014). The origin of oxytocin in the
offspring brain during parturition is under debate. In rodents, the
synthesis of brain oxytocin increase progressively during fetal
development (Altstein and Gainer, 1988). However, the amidate
mature oxytocin is not produced after term (Altstein and Gainer,
1988; Lipari et al., 2001). Thus, it has been proposed that maternal
oxytocin, which is known to cross the placenta (Malek et al., 1996),
is the main source of oxytocin for the fetal brain in rodents (Tyzio
et al., 2006). However, there is evidence of production of oxytocin
by the human fetal brain (Tyzio et al., 2007). With independence of
the origin of oxytocin, an important question that remains to be
determined is whether the reduction of [Cl
]
i
in neurons during
parturition, and the consequent shift of GABA neurotransmission
from excitatory to inhibitory, is altered in absence of labor.
Experiments in mice suggest that the omission of the stages of
labor in elective Cesarean sections may affect brain development of
the offspring. Vaginal delivery, but not Cesarean section, induces
the expression of mitochondrial uncoupling protein 2 (UCP2) in
the newborn hippocampus (Simon-Areces et al., 2012). UCP2 is in-
volved in synaptogenesis, fuel utilization, and mitochondrial bio-
energetics and proliferation. Therefore, the induction of UCP2 by
vaginal delivery may have important consequences for the devel-
oping brain. Indeed, the relevance of the induction of UCP2 in the
newborn hippocampus is suggested by the fact that the pharmaco-
logical inhibition or the genetic ablation of UCP2 lead to dimin-
ished neuronal number, neuronal size, dendritic growth and
synaptogenesis in vitro and impaired spatial memory in adulthood
(Simon-Areces et al., 2012).
Human studies also suggest that the omission of labor may af-
fect brain development in the newborn. The limited available data
indicate that children born from Cesarean delivery at maternal
request, i.e. from Cesarean sections performed when labor is not
initiated, may face more emotional disturbances and sleep prob-
lems at preschool age. In one study, preschool children born by
Cesarean deliveries at maternal request presented significantly
more anxiety/depression, withdrawal and sleep problems. In addi-
tion, they had statistically significantly higher values on internaliz-
ing problems (Kelmanson, 2013). A study of breech-presenting
children did not detect an influence of the mode of delivery on
the incidence of organic brain disorders in later childhood (Preis
et al., 2012). However, some studies suggest that Cesarean surgery
464 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
may increase the risk of autism (Glasson et al., 2004). Autism cases
were more likely to have experienced fetal distress, been delivered
by an elective or emergency Cesarean section, and had an Apgar
score of less than 6 at 1 min (Glasson et al., 2004). Preterm birth
and planned Cesarean section have been also identified as risk fac-
tors for autism and other Pervasive Developmental Disorders in an-
other study (Guinchat et al., 2012). In a meta-analysis, Cesarean
delivery was associated with a 26% increased risk of autism that
did not reach statistical significance (Gardener et al., 2011). Birth
by Cesarean section is also a risk factor for attention deficit hyper-
active disorder (ADHD) in children (Amiri et al., 2012). Unfortu-
nately most studies that review risk for autism or ADHD in
children do not differentiate between elective and urgent
Cesareans.
Concerning the potential consequences that elective Cesarean
section may have for brain development in the newborn it is also
relevant to mention that Cesarean sections may be programmed
earlier than in the due date. This is because the length of human
gestation shows a considerable variation (Jukic et al., 2013), which
can reach even one month. This is a relevant issue, since there is
increasing evidence that birth at 39–41 weeks provides develop-
mental advantages compared to birth at 37–38 weeks. Indeed,
greater gestational age is associated with greater maturity of
movement, better regulation, less excitability and less educational
needs (MacKay et al., 2010; Fink et al., 2012).
A central issue for the main subject of the present paper is the
impact that the omission of labor may have on the process of
mother–child attachment. The lack of the labor-associated nor-
adrenaline peak in the babies born by elective Cesarean section
could hinder the olfactory orientation for the initiation of breast-
feeding. Indeed, as mentioned in Section 2.2, babies born by Cesar-
ean section performed during labor showed a better olfactory
learning during the first hour after birth compared to babies born
by elective Cesarean section without labor (Varendi et al., 2002).
Thus, the omission of labor may potentially affect one of the first
steps in the generation of mother–child attachment. More recent
studies have shown that, depending on the type of delivery, there
are differences in the behavioral responses of the newborns to
maternal separation. A pilot study by our group revealed that neo-
natal crying following brief maternal separation was reduced in
the neonates born by a planned Cesarean section compared to vag-
inally delivered neonates (Olza Fernandez et al., 2013). In addition,
when proximity to the mother was restored, most neonates born
by planned Cesarean section did not cry at all or cried less than
vaginally delivered neonates, which in addition continued crying
for longer time. These different responses could reflect modifica-
tions in early infant attachment behavior (Olza Fernandez et al.,
2013). This different newborn behavior in response to maternal
separation should be understood within the attachment theory
paradigm: lack of response to maternal separation is an early
symptom of un-attachment, which could mean that the generation
of attachment is more difficult for babies born after a planned
Cesarean. Also, the altered response to maternal separation could
indicate an alteration of the stress response, which could be related
to later difficulties with attention or spatial memory processes
similar to the findings in rats described by Simon-Areces et al.
(2012).
The potential negative effects of Cesarean section in children
may be in part due to the impact of Cesarean section on maternal
attitudes and behaviors in relation to their children. Therefore, in
the next section we will review the potential consequences of
Cesarean section for the mother.
4.1.1.2. Consequences for the mother. Cesarean section is associated
with significantly lower maternal levels of catecholamines, adreno-
corticotropic hormone, cortisol, prolactin, corticotropin-releasing
factor, beta-endorphin and oxytocin in plasma (Takagi et al.,
1985; Takeda et al., 1985; Nissen et al., 1996; Vogl et al., 2006). It
might be hypothesized that, in mothers who give birth by planned
Cesarean section, the absence of the neuroendocrine changes asso-
ciated with physiological labor may produce difficulties in the
bonding process and in the recognition by the mother of her child
as her own. This may explain the feeling of estrangement and bond-
ing difficulties that some mothers have described after Cesarean
section (Beck, 2004).
The reduced levels of oxytocin in the cerebrospinal fluid of
mothers in absence of labor (Takagi et al., 1985; Takeda et al.,
1985) may impair maternal adaptations normally induced during
labor and may influence her interactive ability to calm the infant
(Nissen et al., 1998). Although one study found that the mode of
delivery does not affect maternal interaction with the newborn
(Carlander et al., 2010), functional neuroimaging studies have
shown that mothers who give birth by Cesarean have a signifi-
cantly less response in the brain to the cries of their babies than
those who had a vaginal birth (Swain et al., 2008). Lower levels
of brain oxytocin in the mother in absence of labor may also reduce
breastfeeding, which in turn may further affect the process of
mother–child attachment, since breastfeeding increases the intra-
cerebral release of oxytocin in the brain (see Section 3.2). Indeed,
it has been reported that risk of breastfeeding failure is higher after
Cesarean delivery, particularly if primary and scheduled before
39 weeks of gestation (Zanardo et al., 2012, 2013). Several studies
have also investigated the association between type of delivery and
risk of developing postpartum depression. It has been suggested
that Cesarean section may increase the risk of depression (Ryding
et al., 1998). A recent prospective longitudinal study, that recruited
1100 women, reported that those who underwent Cesarean sec-
tion, urgent or elective, had higher scores in the Edinburgh Postna-
tal Depression Scale (EPDS), although six months after birth these
differences were no longer detectable (Rauh et al., 2012). This pos-
sible effect of Cesarean section on maternal depression is also rel-
evant for mother–child attachment, since postpartum depression
causes alterations in the relation of the mother with her baby
(Kingston et al., 2012).
4.1.2. Intrapartum neurohormonal manipulations
4.1.2.1. Synthetic oxytocin. Intrapartum neurohormonal manipula-
tions happen when birth is induced or stimulated with synthetic
oxytocin (Pitocin) or when anesthetic drugs with a central effect
are administered to mothers during childbirth. Other subtle
manipulations happen when birth is disturbed by obstetric inter-
ventions such as amniotomy, lithotomy or unsupportive care but
this have rarely taken into account. Mothers may receive Pitocin
before delivery to induce labor or after delivery to prevent postpar-
tum hemorrhage. In several cases the use of Pitocin is needed for
the health and wellbeing of the mother and child. However, the
use of intrapartum exogenous oxytocin has increased considerably
in recent years (Clark et al., 2009). Possible long-term conse-
quences of the manipulation with synthetic oxytocin in humans
at the time of birth have not been extensively studied, although
some studies suggest that the administration of Pitocin is associ-
ated with an increased number of women abandoning breastfeed-
ing (Ounsted et al., 1978a,b; Wiklund et al., 2009).
The intrapartum administration of oxytocin results in altered
postpartum oxytocin levels in the mother. Thus, higher amount
of exogenous intrapartum oxytocin predicts higher plasma oxyto-
cin levels at 2 months postpartum (Prevost et al., 2014). However,
mothers who receive intrapartum Pitocin, present lower levels of
endogenous oxytocin and increased levels of prolactin two days
after delivery. These changes may be the consequence of a dose-
dependent alteration of the feedback mechanism of both hormones
by Pitocin (Jonas et al., 2009). Thus, it has been proposed that the
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 465
systemic administration of Pitocin, to induce or enhance uterine
contractions, may alter the recycling of oxytocin receptors in the
brain, reducing their availability in the postsynaptic membrane
and therefore decreasing the response to new oxytocin stimuli
(Wahl, 2004). Such a mechanism in the uterus may explain the in-
creased risk of severe maternal bleeding observed when synthetic
oxytocin is administered at birth (Belghiti et al., 2011). Pitocin use
also increases the risk of operative delivery and emergency Cesar-
ean (Buchanan et al., 2012) which in turn may increase the risk of
maternal posttraumatic stress disorder (PTSD) following childbirth
(Beck et al., 2011). The high prevalence of complete or partial PTSD
after childbirth observed in different studies (Ryding et al., 1997;
Wijma et al., 1997; Creedy et al., 2000; Soderquist et al., 2002;
Ayers et al., 2008) raises the question of whether peripartal hor-
monal manipulation alters the maternal neurohormonal milieu at
a time primed for lasting memories.
In addition to the possible effects in the mother, it is also impor-
tant to determine whether intrapartum Pitocin may affect the
newborn. The increase in autism spectrum disorders in the recent
decades and the observation that perinatal factors play a role in
their etiology has led to the hypothesis that oxytocin administra-
tion at birth for labor induction could contribute to the develop-
ment of these diseases (Gardener et al., 2011). Indeed, induction/
augmentation of labor may result in an increased risk of autism,
being this probability increased in male children (Gregory et al.,
2013). Recent studies suggest that Pitocin may interfere with the
initiation of breastfeeding by the newborn (Olza Fernandez et al.,
2012). In a pilot study, we observed a negative association between
oxytocin dose and newborn suckling (Olza Fernandez et al., 2012).
Similar findings have been recently reported in the hour following
birth: fewer prefeeding cues were observed in infants exposed ver-
sus unexposed to Pitocin (Bell et al., 2013). The mechanisms in-
volved in these effects of Pitocin are unknown.
4.1.2.2. Oxytocin receptor antagonists. Oxytocin receptor antago-
nists are frequently used to prevent preterm labor. Atosiban is a
mixed oxytocin/vasopressin V1a receptor antagonist that provides
an effective and more safety therapy than other tocolytic drugs to
delay delivery, being more effective as gestation progresses and the
expression of oxytocin receptor increases (Haas et al., 2012). The
possible long-term consequences of the treatment with Atosiban
for the brain of the mother and the fetus have not been studied.
However, treatment of pregnant rats from days 15 to 20 of gesta-
tion with Atosiban caused impaired lactation, resulting in poor
neonatal survival in offspring (McAnulty and Burns, 2004). In addi-
tion, oxytocin receptor antagonist treatment in pregnant and lac-
tating rats was also shown to increase maternal anxiety levels
and the HPA axis responsiveness, measured as corticosterone lev-
els (Neumann et al., 2000). Furthermore, the intracerebroventricu-
lar administration of an oxytocin receptor antagonist to lactating
dams reduced suckling-evoked activity in different brain regions
related with olfaction, such as the insular cortex, the piriform cor-
tex, the anterior olfactory nucleus and the olfactory tubercle. Other
brain regions in which the oxytocin receptor antagonist reduced
suckling-evoked activity include the prefrontal cortex, the dentate
gyrus and the hippocampal CA1 and CA3 (Febo et al., 2005). There-
fore, this suggests that the antagonism of brain oxytocin receptors
may impair olfactory and cognitive responses elicited by suckling
in the mother brain, which in turn may alter the process of bonding
with the newborn.
Concerning the fetus, since oxytocin is protective for fetal neu-
rons, the antagonism of its receptors may increase the susceptibil-
ity for hypoxic-ischemic insults (Ceanga et al., 2010). Furthermore,
in agreement with the analgesic effect of oxytocin, Atosiban
administration increases pain in rat newborns (Mazzuca et al.,
2011). However, it is unclear whether Atosiban administrated to
the mother may directly affect the newborn. A minimal placental
transfer of Atosiban was detected in a study with eight women
undergoing Cesarean section at term (Valenzuela et al., 1995).
However, the antagonism of oxytocin receptors in the mother
may influence the fetus by indirect mechanisms. Therefore, future
studies are necessary to determine the possible long-term conse-
quences in the infant (Papatsonis et al., 2013).
4.1.2.3. Morphine. In view of the importance of the endogenous
opioid system in modulating oxytocin release, it is of interest to
consider the consequences of the use of exogenous opioids as anal-
gesics in human parturition. Morphine provides women in latent
labor with analgesia and rest and contractions may cease soon
after injection. Morphine is not only used for analgesia, but also
to differentiate true labor from false (Mackeen et al., 2014). The
clinical observation that morphine gives patients rest from con-
tractions and delays childbirth is likely to be due to inhibition of
oxytocin release. As the drug wears off, or as opioid receptors are
down regulated, oxytocin levels rebound significantly and the pa-
tient enters in active labor or return to baseline uterine activity
without labor (Wilson et al., 2010).
A Cochrane systematic review (Anim-Somuah et al., 2011) re-
ported that the use of epidural analgesia in women was associated
with an increase risk of assisted vaginal birth (RR 1.42, 95% CI
1.28–1.57, 23 trials, 7935 women), longer second stage of labor
(MD 13.66 min, 95% CI 6.67–20.66, 13 trials, 4233 women), oxyto-
cin administration (RR 1.19, 95% CI 1.03–1.39, 13 trials, 5815
women) and an increased risk of Cesarean section for fetal distress
(RR 1.43, 95% CI 1.03–1.97, 11 trials, 4816 women). However,
epidural analgesia did not affect the neonatal status, assessed with
the Apgar score. However, further research is needed to evaluate
rare but potential severe adverse effects of epidural analgesia on
women in labor, long-term neonatal outcomes and the impact of
this analgesia on infant–mother attachment and infant
neurodevelopment.
Concerning breastfeeding, although many studies have raised
the question of whether labor epidural analgesia inclusive of opi-
oids has a negative impact on breastfeeding success, the answer re-
mains uncertain. Studies are both conflicting and fraught with
potentially confounding variables such as the mode of delivery,
prior breastfeeding success and parity (Wieczorek et al., 2010;
Szabo, 2013). In the largest study to date, Wilson et al. randomized
1054 patients to various epidural and non-epidural regimens and
did not detect significant differences in breastfeeding success
(Wilson et al., 2010). Further research is needed to elucidate the
specific relationship between neuraxial opioids and breastfeeding
and whether these drugs may act directly on the neonatal brain
to attenuate exhibition of breastfeeding behaviors (Dondi et al.,
1991).
4.2. Preterm delivery
The main factor that interferes with the attachment process in
premature babies is their incomplete brain maturation. Thus, the
full response to olfactory stimuli, which allows the newborn to lo-
cate the nipple, does not take place until about the 29th gestational
week. Also, the response to auditory stimuli, such as the voice of
the mother, cannot be appreciated until near the 32nd gestational
week (Lagercrantz and Changeux, 2010). The bonding process of
the mother and the newborns is also impaired by the separation
of the preterm newborns from their mothers, as a result of their
need of intensive care (see Section 4.3). In consequence, the neuro-
endocrine changes that are associated with mother–child syn-
chrony are not elicited in premature babies. This may have
consequences not only for the preterm newborn but also for the
parents. Preterm birth and hospitalization of the preterm infant
466 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
have been associated with a high incidence of parental PTSD that
may persist even eighteen months after birth and impair parent–
infant interaction (Pierrehumbert et al., 2003; Forcada-Guex
et al., 2006; Shaw et al., 2009). Thus it seems crucial to include
perinatal specialized psychological care in the entire context of
prematurity for these families (Hatters Friedman et al., 2013) and
to promote actions to restore contact of the newborn with the
mother or father, such as Kangaroo Mother care (KMC) method
and the Newborn Individualized Developmental Care and Assess-
ment Program (NIDCAP). The KMC implies continuous skin-to-skin
contact between the mother and the infant, exclusive breastfeed-
ing and early home discharge in the kangaroo position (Charpak
et al., 2005). It has been reported that KMC enhances bonding
and attachment, reduces maternal postpartum depression symp-
toms, enhances infant physiologic stability, reduces pain, increases
parental sensitivity to infant cues, contributes to the establishment
and longer duration of breastfeeding and has positive effects on in-
fant development and infant/parent interaction (Nyqvist et al.,
2010). The NIDCAP also mimics the intrauterine environment, min-
imizing environmental stress and promoting physiological individ-
ualized care (Als et al., 1996).
4.3. Mother–infant postpartum separation
Sometimes, as consequence of maternal or neonatal pathology,
or due to practices without scientific evidence, such as limiting
access to parents to neonatal units or avoiding the postpartum
mother–infant skin-to-skin contact, separation occurs between
the mother and the newborn immediately after childbirth.
Mother–infant postpartum interaction modifies oxytocin levels in
both the mother and the newborn brain (Kendrick, 2000). There-
fore, the deprivation of the physical and tactile stimuli provided
by the mother may have potential long-term consequences for
the newborn. Although this question has not been adequately
studied in humans, it is known that the deprivation of contact with
the mother results in long-term affective and cognitive alterations
in rodents (Meaney et al., 2002; Nagasawa et al., 2012; Marco et al.,
2013).
Maternal separation in rodents not only affects the newborns
but also the behavior of the mother. In rats, maternal separation re-
duces maternal care (Aguggia et al., 2013). In addition, long periods
of mother–infant separation during the postnatal period inhibit
subsequent maternal behavior, probably though oxytocin receptor
modulation in the brain (Boccia and Pedersen, 2001). In humans,
separation and swaddling of the baby at birth impairs the interac-
tion with the mother during breastfeeding and also affects the
behavior of the mother with her child (Dumas et al., 2013). The ef-
fects of the early maternal separation are durable. A study reported
that, regardless of other variables, when newborns spend the first
two hours separated from their mothers there was a worse
mother–baby interaction one year after birth (Bystrova et al.,
2009). The stress of prolonged mother–infant separation is also
associated with reduced maternal sensitivity and more negative
patterns of mothering throughout the first 3 years of life (NICHD
Early Child Care Research Network, 1999).
4.4. Bottle-feeding versus breastfeeding
Neuroendocrine setting of bottle-feeding may be very different
from lactation. In bottle-feeding, the absence of the increased re-
lease of oxytocin and prolactin in the mother induced by breast-
feeding may affect the bonding process with her newborn. This
has led some authors to hypothesize that, for the maternal brain,
not breastfeeding may mean the ‘death’ of the newborn. Therefore,
the brain might be involved in a postpartum mourning state that
may explain the higher incidence of postpartum depression in
the case of bottle-feeding (Gallup et al., 2010). Indeed, a lack of
brain activation in response to the crying of the newborn, similar
to what it has been described after elective Cesarean sections,
has been reported in mothers that use bottle-feeding (Swain
et al., 2008; Kim et al., 2011). Long term consequences of these
differences are still unknown. Duration of breastfeeding has been
inversely associated with risk of subsequent maternal neglect in
a large 15-year longitudinal study of over 7000 mother–infant
dyads (Strathearn et al., 2009b). In addition, a significant correla-
tion between stress, dysphoric moods and decreased levels of
interferon-
c
have been detected in formula feeder mothers, an ef-
fect consistent with depression of cellular immunity (Groer and
Davis, 2006). Furthermore, recent studies suggest that breastfeed-
ing reduces women’s risk of Alzheimer’s disease and that women
with shorter breastfeeding duration have increased risk of Alzhei-
mer’s disease (Fox et al., 2013).
4.5. Summation of effects
Unfortunately many mother infant dyads suffer not one but
many of these peripartum disruptions. For instance, prematurity
can be associated to betamethasone and Atosiban exposure in
uterus, followed by an emergency Cesarean birth, maternal infant
separation, prolonged hospitalization and formula feeding. In some
countries maternal infant abandonment may be followed by for-
mula feeding and institutionalization until 10 months of age with-
out a primary caregiver (Gribble, 2006). The consequences of the
accumulative effects of these events, which impact on the neuro-
endocrine homeostasis of the baby, are unknown. However, it
has been reported that birth complications and early child rejec-
tion increases the risk of violent behavior in adulthood (Raine
et al., 1994, 1997). It is unknown if the summation of potential
etiopathogenic neurohormonal scenarios during labor and the
early postpartum period may represent an increased risk for other
neurobiological disorders that affect systems and functions regu-
lated by neurohormones such as attachment disorders, childhood
anxiety disorders, early feeding disorders, changes in sexual behav-
ior in adulthood or primary enuresis.
5. Conclusions
Childbirth can be considered as a neurohormonal event where
both maternal and fetal brains participate and where a specific
neurohormonal scenario is settled. The studies reviewed in this
paper suggest that hormonal changes in the maternal and the fe-
tal/newborn brains during parturition, the immediate postpartum
period and lactation are involved in the generation of mother–
child attachment in humans. Several hormones seem to play a
key role in the mechanisms of initiation and consolidation of this
attachment. However, our knowledge on these mechanisms is
still incomplete. Little is known on the neurohormonal changes
during intrapartum and we ignore the neurobiological and neuro-
endocrine changes that may occur in the maternal and the fetal
brain as the fetus goes down the birth channel. Also, we need
to increase our knowledge on the mechanisms that initiate
attachment behavior between the mother and the newborn
immediately postpartum. This knowledge will allow the imple-
mentation of adequate actions to minimize the negative effects
of peripartum disruptions. For this it is essential to obtain a sys-
tematic recollection of data regarding any peripartum attachment
disruption in the medical records, such as the time, dose and
duration of synthetic oxytocin exposure or of maternal infant
separation. More investigation is needed to develop scores to as-
sess peripartum disruptions and identify high risk neonates and
dyads.
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 467
The limited available evidence suggests that the disruption of
the neurohormonal process of childbirth in humans may increase
the risk of developing mental, learning, attachment and personality
disorders later in life. Therefore, several actions to counteract the
effects of birth interventions disrupting peripartal neuroendocrine
events have been proposed, including strategies that maximizie
skin-to-skin contact, carrying the child frequently, providing mas-
sage, co-sleeping and co-bathing (see also Section 4.2). In general,
it would be convenient to educate both professionals and parents
on the critical issues that take place in the central nervous system
at delivery or during perinatal period as well as to promote phys-
iological care during childbirth, accommodating hospital routines
to newborns basic biological needs (Bergman and Bergman,
2013), minimizing pharmacological interventions that may disrupt
the neuroendocrine cascade involved in mother–child attachment,
performing Cesarean sections as physiological as possible (Smith
et al., 2008) and promoting strategies to decrease the alarmingly
high birth rate by Cesarean sections, many of them unnecessary
(Young, 2011). Routine mother infant separation should be
avoided. Instead, skin-to-skin contact with the mother for the first
24 h should be provided when possible to keep uninterrupted ac-
cess to the breast. When breastfeeding is not possible, or is not
the preferred option, it is recommended to bottle feed mimicking
breastfeeding, holding the child in a breastfeeding position with
the child skin-to-skin with the mother bare breasts, using a
slow-flow bottle teat and changing sides while bottle feeding
(Gribble, 2006). If maternal illness causes infant separation, skin
to skin contact with the father or other relatives should be pro-
vided and intensive breastfeeding should be promoted after re-
union with the mother to restore brain oxytocin levels. Finally,
screening for perinatal mental health disorders seems to be critical
to optimize maternal well-being and attachment. Perinatal and in-
fant mental health services should be provided to all families who
experience traumatic childbirth and or prematurity.
Acknowledgments
Grant support from the Fondo de Investigaciones Sanitarias
(PI10/00791) and Ministerio de Economía y Competitividad, Spain
(BFU2011-30217-C03-01).
References
Abel, R.A., Ronca, A.E., Alberts, J.R., 1998. Perinatal stimulation facilitates suckling
onset in newborn rats. Dev. Psychobiol. 32, 91–99.
Aguggia, J.P., Suarez, M.M., Rivarola, M.A., 2013. Early maternal separation:
neurobehavioral consequences in mother rats. Behav. Brain Res. 248, 25–31.
Akbari, E.M., Chatterjee, D., Lévy, F., Fleming, A.S., 2007. Experience-dependent cell
survival in the maternal rat brain. Behav. Neurosci. 121, 1001–1011.
Als, H., Duffy, F.H., McAnulty, G.B., 1996. Effectiveness of individualized
neurodevelopmental care in the newborn intensive care unit (NICU). Acta
Paediatr. Suppl. 416, 21–30.
Altemus, M., Deuster, P.A., Galliven, E., Carter, C.S., Gold, P.W., 1995. Suppression of
hypothalmic-pituitary-adrenal axis responses to stress in lactating women. J.
Clin. Endocrinol. Metab. 80, 2954–2959.
Altstein, M., Gainer, H., 1988. Differential biosynthesis and posttranslational
processing of vasopressin and oxytocin in rat brain during embryonic and
postnatal development. J. Neurosci. 8, 3967–3977.
Amico, J.A., Finley, B.E., 1986. Breast stimulation in cycling women, pregnant
women and a woman with induced lactation: pattern of release of oxytocin,
prolactin and luteinizing hormone. Clin. Endocrinol. (Oxf) 25, 97–106.
Amico, J.A., Thomas, A., Hollingshead, D.J., 1997. The duration of estradiol and
progesterone exposure prior to progesterone withdrawal regulates oxytocin
mRNA levels in the paraventricular nucleus of the rat. Endocr. Res. 23, 141–156.
Amiri, S., Malek, A., Sadegfard, M., Abdi, S., 2012. Pregnancy-related maternal risk
factors of attention-deficit hyperactivity disorder: a case-control study. ISRN
Pediatr. 2012, 458064.
Anim-Somuah, M., Smyth, R.M., Jones, L., 2011. Epidural versus non-epidural or no
analgesia in labour. Cochrane Database Syst. Rev. 12, CD000331.
Avinun, R., Ebstein, R.P., Knafo, A., 2012. Human maternal behaviour is associated
with arginine vasopressin receptor 1A gene. Biol. Lett. 8, 894–896.
Ayers, S., Joseph, S., McKenzie-McHarg, K., Slade, P., Wijma, K., 2008. Post-traumatic
stress disorder following childbirth: Current issues and recommendations for
future research. J. Psychosom. Obste. Gynaecol. 29, 240–250.
Bamberg, C., Rademacher, G., Guttler, F., Teichgraber, U., Cremer, M., Buhrer, C.,
Dudenhausen, J.W., 2012. Human birth observed in real-time open magnetic
resonance imaging. Am. J. Obstet. Gynecol. 206, 505.e1-6.
Baumgartner, T., Heinrichs, M., Vonlanthen, A., Fischbacher, U., Fehr, E., 2008.
Oxytocin shapes the neural circuitry of trust and trust adaptation in humans.
Neuron 58, 639–650.
Bealer, S.L., Crowley, W.R., 2001. Histaminergic control of oxytocin release in the
paraventricular nucleus during lactation in rats. Exp. Neurol. 171, 317–322.
Beck, C.T., 2004. Post-traumatic stress disorder due to childbirth: the aftermath.
Nurs. Res. 53, 216–224.
Beck, C.T., Gable, R.K., Sakala, C., Declercq, E.R., 2011. Posttraumatic stress disorder
in new mothers: results from a two-stage U.S. National survey. Birth 38, 216–
227.
Belghiti, J., Kayem, G., Dupont, C., Rudigoz, R.C., Bouvier-Colle, M.H., Deneux-
Tharaux, C., 2011. Oxytocin during labour and risk of severe postpartum
haemorrhage: a population-based, cohort-nested case-control study. BMJ Open
1, e000514.
Bell, A.F., White-Traut, R., Rankin, K., 2013. Fetal exposure to synthetic oxytocin and
the relationship with prefeeding cues within one hour postbirth. Early Hum.
Dev. 89, 137–143.
Bergman, J., Bergman, N., 2013. Whose choice? advocating birthing practices
according to baby’s biological needs. J. Perinatal Edu. 22, 8–13.
Bisceglia, R., Jenkins, J.M., Wigg, K.G., O’Connor, T.G., Moran, G., Barr, C.L., 2012.
Arginine vasopressin 1a receptor gene and maternal behavior: evidence of
association and moderation. Genes Brain. Behav. 11, 262–268.
Boccia, M.L., Pedersen, C.A., 2001. Brief vs. long maternal separations in infancy:
contrasting relationships with adult maternal behavior and lactation levels of
aggression and anxiety. Psychoneuroendocrinology 26, 657–672.
Bosch, O.J., 2011. Maternal nurturing is dependent on her innate anxiety: the
behavioral roles of brain oxytocin and vasopressin. Horm. Behav. 59, 202–212.
Bosch, O.J., Neumann, I.D., 2008. Brain vasopressin is an important regulator of
maternal behavior independent of dams’ trait anxiety. Proc. Natl. Acad. Sci. USA
105, 17139–17144.
Bosch, O.J., Neumann, I.D., 2012. Both oxytocin and vasopressin are mediators of
maternal care and aggression in rodents: from central release to sites of action.
Horm. Behav. 61, 293–303.
Bowlby, J., 1978. Attachment theory and its therapeutic implications. Adolesc.
Psychiatry 6, 5–33.
Brancato, R.M., Church, S., Stone, P.W., 2008. A meta-analysis of passive descent
versus immediate pushing in nulliparous women with epidural analgesia in the
second stage of labor. J. Obstet. Gynecol. Neonatal. Nurs. 37, 4–12.
Brunton, P.J., Russell, J.A., 2008. The expectant brain: adapting for motherhood. Nat.
Rev. Neurosci. 9, 11–25.
Brunton, P.J., Russell, J.A., 2010. Endocrine induced changes in brain function during
pregnancy. Brain Res. 1364, 198–215.
Brunton, P.J., Russell, J.A., Douglas, A.J., 2008. Adaptive responses of the maternal
hypothalamic-pituitary-adrenal axis during pregnancy and lactation. J.
Neuroendocrinol. 20, 764–776.
Brunton, P.J., Bales, J., Russell, J.A., 2012. Allopregnanolone and induction of
endogenous opioid inhibition of oxytocin responses to immune stress in
pregnant rats. J. Neuroendocrinol. 24, 690–700.
Brunton, P.J., Russell, J.A., Hirst, J.J., 2014. Allopregnanolone in the brain: protecting
pregnancy and birth outcomes. Prog. Neurobiol. 113, 106–136.
Brus, M., Meurisse, M., Franceschini, I., Keller, M., Lévy, F., 2010. Evidence for cell
proliferation in the sheep brain and its down-regulation by parturition and
interactions with the young. Horm. Behav. 58, 737–746.
Brussaard, A.B., Kits, K.S., Baker, R.E., Willems, W.P., Leyting-Vermeulen, J.W., Voorn,
P., Herbison, A.E., 1997. Plasticity in fast synaptic inhibition of adult oxytocin
neurons caused by switch in GABA(A) receptor subunit expression. Neuron 19,
1103–1114.
Brussaard, A.B., Devay, P., Leyting-Vermeulen, J.L., Kits, K.S., 1999. Changes in
properties and neurosteroid regulation of GABAergic synapses in the supraoptic
nucleus during the mammalian female reproductive cycle. J. Physiol. 516, 513–
524.
Buchanan, S.L., Patterson, J.A., Roberts, C.L., Morris, J.M., Ford, J.B., 2012. Trends and
morbidity associated with oxytocin use in labour in nulliparas at term. Aust. N.
Z. J. Obstet. Gynaecol. 52, 173–178.
Bystrova, K., Widström, A.M., Matthiesen, A.S., Ransjö-Arvidson, A.B., Welles-
Nyström, B., Wassberg, C., Vorontsov, I., Uvnäs-Moberg, K., 2003. Skin-to-skin
contact may reduce negative consequences of ‘‘the stress of being born’’: a
study on temperature in newborn infants, subjected to different ward routines
in St. Petersburg. Acta Paediatr. 92, 320–326.
Bystrova, K., Ivanova, V., Edhborg, M., Matthiesen, A.S., Ransjo-Arvidson, A.B.,
Mukhamedrakhimov, R., Widstrom, A.M., 2009. Early contact versus separation:
effects on mother–infant interaction one year later. Birth 36, 97–109.
Callister, L.C., 2004. Making meaning: women’s birth narratives. J. Obstet. Gynecol.
Neonatal. Nurs. 33, 508–518.
Callister, L.C., Semenic, S., Foster, J.C., 1999. Cultural and spiritual meanings of
childbirth. Orthodox jewish and mormon women. J. Holist. Nurs. 17, 280–
295.
Campbell, E.A., Linton, E.A., Wolfe, C.D., Scraggs, P.R., Jones, M.T., Lowry, P.J., 1987.
Plasma corticotropin-releasing hormone concentrations during pregnancy
and parturition. J. Clin. Endocrinol. Metab. 64, 1054–1059.
468 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
Carlander, A.K., Edman, G., Christensson, K., Andolf, E., Wiklund, I., 2010. Contact
between mother, child and partner and attitudes towards breastfeeding in
relation to mode of delivery. Sex. Reprod. Healthc. 1, 27–34.
Carter, C.S., 2014. Oxytocin pathways and the evolution of human behavior. Annu.
Rev. Psychol. 65, 17–39.
Carter, C.S., Altemus, M., 1997. Integrative functions of lactational hormones in
social behavior and stress management. Ann. N. Y. Acad. Sci. 807, 164–174.
Caughey, S.D., Klampfl, S.M., Bishop, V.R., Pfoertsch, J., Neumann, I.D., Bosch, O.J.,
Meddle, S.L., 2011. Changes in the intensity of maternal aggression and central
oxytocin and vasopressin V1a receptors across the peripartum period in the rat.
J. Neuroendocrinol. 23, 1113–1124.
Ceanga, M., Spataru, A., Zagrean, A.M., 2010. Oxytocin is neuroprotective against
oxygen–glucose deprivation and reoxygenation in immature hippocampal
cultures. Neurosci. Lett. 477, 15–18.
Charpak, N., Ruiz, J.G., Zupan, J., Cattaneo, A., Figueroa, Z., Tessier, R., Cristo, M.,
Anderson, G., Ludington, S., Mendoza, S., Mokhachane, M., Worku, B., 2005.
Kangaroo mother care: 25 years after. Acta Paediatr. 94, 514–522.
Chen, D.C., Nommsen-Rivers, L., Dewey, K.G., Lonnerdal, B., 1998. Stress during labor
and delivery and early lactation performance. Am. J. Clin. Nutr. 68, 335–344.
Cherubini, E., Gaiarsa, J.L., Ben-Ari, Y., 1991. GABA: an excitatory transmitter in early
postnatal life. Trends Neurosci. 14, 515–519.
Chiossi, G., Lai, Y., Landon, M.B., Spong, C.Y., Rouse, D.J., Varner, M.W., Eunice
Kennedy Shriver National Institute of Child Health and Human Development
(NICHD) Maternal-Fetal Medicine Units (MFMU) Network, 2013. Timing of
delivery and adverse outcomes in term singleton repeat cesarean deliveries.
Obstet. Gynecol. 121, 561–569.
Christensson, K., Cabrera, T., Christensson, E., Uvnas-Moberg, K., Winberg, J., 1995.
Separation distress call in the human neonate in the absence of maternal body
contact. Acta Paediatr. 84, 468–473.
Clark, S.L., Simpson, K.R., Knox, G.E., Garite, T.J., 2009. Oxytocin: new perspectives
on an old drug. Am. J. Obstet. Gynecol. 35, e1–e6.
Costa, A., De Filippis, V., Voglino, M., Giraudi, G., Massobrio, M., Benedetto, C.,
Marozio, L., Gallo, M., Molina, G., Fabris, C., 1988. Adrenocorticotropic hormone
and catecholamines in maternal, umbilical and neonatal plasma in relation to
vaginal delivery. J. Endocrinol. Invest. 11, 703–709.
Creedy, D.K., Shochet, I.M., Horsfall, J., 2000. Childbirth and the development of
acute trauma symptoms: incidence and contributing factors. Birth 27, 104–111.
Crowley, R.S., Insel, T.R., O’Keefe, J.A., Kim, N.B., Amico, J.A., 1995. Increased
accumulation of oxytocin messenger ribonucleic acid in the hypothalamus of
the female rat: induction by long term estradiol and progesterone
administration and subsequent progesterone withdrawal. Endocrinology 136,
224–231.
Da Costa, A.P., Guevara-Guzman, R.G., Ohkura, S., Goode, J.A., Kendrick, K.M., 1996.
The role of oxytocin release in the paraventricular nucleus in the control
of maternal behaviour in the sheep. J. Neuroendocrinol. 8, 163–177.
Dahlen, H.G., Kennedy, H.P., Anderson, C.M., Bell, A.F., Clark, A., Foureur, M., Downe,
S., 2013. The EPIIC hypothesis: intrapartum effects on the neonatal epigenome
and consequent health outcomes. Med. Hypotheses 80, 656–662.
Darnaudery, M., Perez-Martin, M., Del Favero, F., Gomez-Roldan, C., Garcia-Segura,
L.M., Maccari, S., 2007. Early motherhood in rats is associated with a
modification of hippocampal function. Psychoneuroendocrinology 32, 803–812.
Dawood, M.Y., Khan-Dawood, F.S., Wahi, R.S., Fuchs, F., 1981. Oxytocin release and
plasma anterior pituitary and gonadal hormones in women during lactation. J.
Clin. Endocrinol. Metab. 52, 678–683.
Deoni, S.C., Dean 3rd., D.C., Piryatinksy, I., O’Muircheartaigh, J., Waskiewicz, N.,
Lehman, K., Dirks, H., 2013. Breastfeeding and early white matter development:
a cross-sectional study. Neuroimage 15, 77–86.
Detwyler, K., 1995. A time to wean. In: Stuar-Macadam, P., Detwyler, K. (Eds.),
Breastfeeding Biocultural Perspectives. Aldine De Gruyter, New York, pp. 39–73.
Dondi, D., Maggi, R., Panerai, A.E., Piva, F., Limonta, P., 1991. Hypothalamic
opiatergic tone during pregnancy, parturition and lactation in the rat.
Neuroendocrinology 53, 460–466.
Donner, N., Bredewold, R., Maloumby, R., Neumann, I.D., 2007. Chronic intracerebral
prolactin attenuates neuronal stress circuitries in virgin rats. Eur. J. Neurosci.
25, 1804–1814.
Doucet, S., Soussignan, R., Sagot, P., Schaal, B., 2007. The ‘‘smellscape’’ of mother’s
breast: effects of odor masking and selective unmasking on neonatal arousal,
oral, and visual responses. Dev. Psychobiol. 49, 129–138.
Douglas, A.J., 2005. Central noradrenergic mechanisms underlying acute stress
responses of the Hypothalamo-pituitary-adrenal axis: adaptations through
pregnancy and lactation. Stress 8, 5–18.
Douglas, A.J., Dye, S., Leng, G., Russell, J.A., Bicknell, R.J., 1993. Endogenous opioid
regulation of oxytocin secretion through pregnancy in the rat. J.
Neuroendocrinol. 5, 307–314.
Douglas, A.J., Neumann, I., Meeren, H.K., Leng, G., Johnstone, L.E., Munro, G., Russell,
J.A., 1995. Central endogenous opioid inhibition of supraoptic oxytocin neurons
in pregnant rats. J. Neurosci. 15, 5049–5057.
Dumas, L., Lepage, M., Bystrova, K., Matthiesen, A.S., Welles-Nystrom, B., Widstrom,
A.M., 2013. Influence of skin-to-skin contact and rooming-in on early mother-
infant interaction: a randomized controlled trial. Clin. Nurs. Res. 22, 310–336.
Duna, J., Richards, M., 1977. Observation on the developing relationship between
mother and baby in the neonatal period. In: Scaefeer, H.R. (Ed.), Studies in
Mother–Infant Interaction. Academic Press, New York, pp. 427–455.
Erlandsson, K., Dsilna, A., Fagerberg, I., Christensson, K., 2007. Skin-to-skin care with
the father after cesarean birth and its effect on newborn crying and prefeeding
behavior. Birth 34, 105–114.
Febo, M., Numan, M., Ferris, C.F., 2005. Functional magnetic resonance imaging
shows oxytocin activates brain regions associated with mother-pup bonding
during suckling. J. Neurosci. 25, 11637–11644.
Feldman, R., Gordon, I., Zagoory-Sharon, O., 2010a. The cross-generation
transmission of oxytocin in humans. Horm. Behav. 58, 669–676.
Feldman, R., Gordon, I., Schneiderman, I., Weissman, O., Zagoory-Sharon, O., 2010b.
Natural variations in maternal and paternal care are associated with systematic
changes in oxytocin following parent-infant contact.
Psychoneuroendocrinology 35, 1133–1141.
Ferber, S.G., Makhoul, I.R., 2004. The effect of skin-to-skin contact (kangaroo care)
shortly after birth on the neurobehavioral responses of the term newborn: a
randomized, controlled trial. Pediatrics 113, 858–865.
Fink, N.S., Tronick, E., Olson, K., Lester, B., 2012. Healthy newborns’ neurobehavior:
norms and relations to medical and demographic factors. J. Pediatr. 161, 1073–
1079.
Fleming, A.S., Steiner, M., Anderson, V., 1987. Hormonal and attitudinal correlates of
maternal behaviour during the early postpartum period in first-time mothers. J.
Reprod. Infant Psychol. 5, 193–205.
Fleming, A.S., Steiner, M., Corter, C., 1997. Cortisol, hedonics, and maternal
responsiveness in human mothers. Horm. Behav. 32, 85–98.
Forcada-Guex, M., Pierrehumbert, B., Borghini, A., Moessinger, A., Muller-Nix, C.,
2006. Early dyadic patterns of mother-infant interactions and outcomes of
prematurity at 18 months. Pediatrics 118, e107–e114.
Fox, M., Berzuini, C., Knapp, L.A., 2013. Maternal breastfeeding history and
Alzheimer’s disease risk. J. Alzheimers Dis. 37, 809–821.
Furuta, M., Bridges, R.S., 2009. Effects of maternal behavior induction and pup
exposure on neurogenesis in adult, virgin female rats. Brain Res. Bull. 80, 408–
413.
Galea, L.A., Ormerod, B.K., Sampath, S., Kostaras, X., Wilkie, D.M., Phelps, M.T., 2000.
Spatial working memory and hippocampal size across pregnancy in rats. Horm.
Behav. 37, 86–95.
Gallup Jr, G.G., Nathan Pipitone, R., Carrone, K.J., Leadholm, K.L., 2010. Bottle feeding
simulates child loss: postpartum depression and evolutionary medicine. Med.
Hypotheses 74, 174–176.
Garcia-Segura, L.M., 2009. Hormones and Brain Plasticity. Oxford University Press,
New York.
Gardener, H., Spiegelman, D., Buka, S.L., 2011. Perinatal and neonatal risk factors for
autism: a comprehensive meta-analysis. Pediatrics 128, 344–355.
Gennser, G., Ohrlander, S., Eneroth, P., 1977. Fetal cortisol and the initiation of
labour in the human. Ciba Found. Symp. 47, 401–426.
Gibbons, L., Belizan, J.M., Lauer, J.A., Betran, A.P., Merialdi, M., Althabe, F., 2012.
Inequities in the use of cesarean section deliveries in the world. Am. J. Obstet.
Gynecol. 206, 331.
Gionet, L., 2013. Breastfeeding trends in Canada. Health at a Glance. November.
Statistics Canada, Catalogue No. 82-624-X.
Gitau, R., Menson, E., Pickles, V., Fisk, N.M., Glover, V., MacLachlan, N., 2001.
Umbilical cortisol levels as an indicator of the fetal stress response to assisted
vaginal delivery. Eur. J. Obstet. Gynecol. Reprod. Biol. 98, 14–17.
Glasson, E.J., Bower, C., Petterson, B., de Klerk, N., Chaney, G., Hallmayer, J.F., 2004.
Perinatal factors and the development of autism: a population study. Arch. Gen.
Psychiatry 61, 618–627.
Gordon, I., Zagoory-Sharon, O., Leckman, J.F., Feldman, R., 2010. Prolactin, oxytocin,
and the development of paternal behavior across the first six months of
fatherhood. Horm. Behav. 58, 513–518.
Graham, M.D., Rees, S.L., Steiner, M., Fleming, A.S., 2006. The effects of
adrenalectomy and corticosterone replacement on maternal memory in
postpartum rats. Horm. Behav. 49, 353–361.
Green, A.D., Galea, L.A., 2008. Adult hippocampal cell proliferation is suppressed
with estrogen withdrawal after a hormone-simulated pregnancy. Horm. Behav.
54, 203–211.
Gregory, S.G., Anthopolos, R., Osgood, C.E., Grotegut, C.A., Miranda, M.L., 2013.
Association of autism with induced or augmented childbirth in North Carolina
birth record (1990–1998) and education research (1997–2007) databases. JAMA
Pediatr. 167, 959–966.
Gribble, K.D., 2006. Mental health, attachment and breastfeeding: implications for
adopted children and their mothers. Int. Breastfeed J. 1, 5.
Groer, M.W., Davis, M.W., 2006. Cytokines, infections, stress, and dysphoric moods
in breastfeeders and formula feeders. J. Obstet. Gynecol. Neonatal. Nurs. 35,
599–607.
Guinchat, V., Thorsen, P., Laurent, C., Cans, C., Bodeau, N., Cohen, D., 2012. Pre-,
peri- and neonatal risk factors for autism. Acta Obstet. Gynecol. Scand. 91,
287–300.
Haas, D.M., Caldwell, D.M., Kirkpatrick, P., McIntosh, J.J., Welton, N.J., 2012. Tocolytic
therapy for preterm delivery: systematic review and network meta-analysis.
BMJ 345, e6226.
Hallowell, S.G., Spatz, D.L., 2012. The relationship of brain development and
breastfeeding in the late-preterm infant. J. Pediatr. Nurs. 27, 154–162.
Hamilton, B.E., Hoyert, D.L., Martin, J.A., Strobino, D.M., Guyer, B., 2013. Annual
summary of vital statistics: 2010–2011. Pediatrics 131, 548–558.
Handlin, L., Jonas, W., Petersson, M., Ejdebäck, M., Ransjö-Arvidson, A.B., Nissen, E.,
Uvnäs-Moberg, K., 2009. Effects of sucking and skin-to-skin contact on maternal
ACTH and cortisol levels during the second day postpartum-influence of
epidural analgesia and oxytocin in the perinatal period. Breastfeed Med. 4, 207–
220.
Hanson, L., 2009. Second-stage labor care: challenges in spontaneous bearing down.
J. Perinat. Neonatal Nurs. 23, 31–39.
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 469
Hatters Friedman, S., Kessler, A., Nagle Yang, S., Parsons, S., Friedman, H., Martin, R.J.,
2013. Delivering perinatal psychiatric services in the neonatal intensive care
unit. Acta Paediatr. 102, e392–e397.
Heinrichs, M., Meinlschmidt, G., Neumann, I., Wagner, S., Kirschbaum, C., Ehlert, U.,
Hellhammer, D.H., 2001. Effects of suckling on hypothalamic-pituitary-adrenal
axis responses to psychosocial stress in postpartum lactating women. J. Clin.
Endocrinol. Metab. 86, 4798–4804.
Heinrichs, M., Neumann, I., Ehlert, U., 2002. Lactation and stress: protective effects
of breast-feeding in humans. Stress 5, 195–203.
Heinrichs, M., von Dawans, B., Domes, G., 2009. Oxytocin, vasopressin, and human
social behavior. Front. Neuroendocrinol. 30, 548–557.
Isaacs, E.B., Fischl, B.R., Quinn, B.T., Chong, W.K., Gadian, D.G., Lucas, A., 2010. Impact
of breast milk on intelligence quotient, brain size, and white matter
development. Pediatr. Res. 67, 357–362.
Ishak, W.W., Kahloon, M., Fakhry, H., 2011. Oxytocin role in enhancing well-being: a
literature review. J. Affect. Disord. 130, 1–9.
Jirikowski, G.F., Caldwell, J.D., Pilgrim, C., Stumpf, W.E., Pedersen, C.A., 1989.
Changes in immunostaining for oxytocin in the forebrain of the female rat
during late pregnancy, parturition and early lactation. Cell Tissue Res. 256, 411–
417.
Johnstone, H.A., Wigger, A., Douglas, A.J., Neumann, I.D., Landgraf, R., Seckl, J.R.,
Russell, J.A., 2000. Attenuation of hypothalamic-pituitary-adrenal axis stress
responses in late pregnancy: changes in feedforward and feedback mechanisms.
J. Neuroendocrinol. 12, 811–822.
Jonas, K., Johansson, L.M., Nissen, E., Ejdeback, M., Ransjo-Arvidson, A.B., Uvnas-
Moberg, K., 2009. Effects of intrapartum oxytocin administration and epidural
analgesia on the concentration of plasma oxytocin and prolactin, in response to
suckling during the second day postpartum. Breastfeed Med. 4, 71–82.
Jukic, A.M., Baird, D.D., Weinberg, C.R., McConnaughey, D.R., Wilcox, A.J., 2013.
Length of human pregnancy and contributors to its natural variation. Hum.
Reprod. 28, 2848–2855.
Julvez, J., Guxens, M., Carsin, A.E., Forns, J., Mendez, M., Turner, M.C., Sunyer, J., 2014.
A cohort study on full breastfeeding and child neuropsychological
development: the role of maternal social, psychological, and nutritional
factors. Dev. Med. Child Neurol. 56, 148–156.
Kafouri, S., Kramer, M., Leonard, G., Perron, M., Pike, B., Richer, L., Paus, T., 2013.
Breastfeeding and brain structure in adolescence. Int. J. Epidemiol. 42, 150–
159.
Kapellou, O., 2011. Effect of caesarean section on brain maturation. Acta Paediatr.
100, 1416–1422.
Kelmanson, I., 2013. Emotional and behavioural features of preschool children born
by caesarean deliveries at maternal request. Eur. J. Dev. Psychol. 10, 676–690.
Kendrick, K.M., 2000. Oxytocin, motherhood and bonding. Exp. Physiol. 85, 111S–
124S.
Kendrick, K.M., Keverne, E.B., 1991. Importance of progesterone and estrogen
priming for the induction of maternal behavior by vaginocervical stimulation in
sheep. Effects of maternal experience. Physiol. Behav. 49, 745–750.
Kendrick, K.M., Keverne, E.B., Chapman, C., Baldwin, B.A., 1988. Microdialysis
measurement of oxytocin, aspartate, gamma aminobutyric acid and glutamate
release from the olfactory bulb of sheep during vaginocervical stimulation.
Brain Res. 442, 171–177.
Kendrick, K.M., Keverne, E.B., Hinton, M.R., Goode, J.A., 1991a. Cerebrospinal fluid
and plasma concentrations of oxytocin and vasopressin during parturition and
vaginocervical stimulation in the sheep. Brain Res. Bull. 26, 803–807.
Kendrick, K.M., Lévy, F., Keverne, E.B., 1991b. Importance of vaginocervical
stimulation for the formation of maternal bonding in primiparous and
multiparous parturient ewes. Physiol. Behav. 50, 595–600.
Kendrick, K.M., Da Costa, A.P., Broad, K.D., Ohkura, S., Guevara, R., Lévy, F., Keverne,
E.B., 1997. Neural control of maternal behaviour and olfactory recognition of
offspring. Brain Res. Bull. 44, 383–395.
Keri, S., Kiss, I., 2010. Oxytocin response in a trust game and habituation of arousal.
Physiol. Behav. 102, 221–224.
Keverne, E.B., 1988. Central mechanisms underlying the neural and neuroendocrine
determinants of maternal behaviour. Psychoneuroendocrinology 13, 127–141.
Keverne, E.B., Lévy, F., Poindron, P., Lindsay, D., 1983. Vaginal stimulation. An
important determinant of maternal bonding in sheep. Science 219, 81–83.
Keyser-Marcus, L., Stafisso-Sandoz, G., Gerecke, K., Jasnow, A., Nightingale, L.,
Lambert, K.G., Kinsley, C.H., 2001. Alterations of medial preoptic area neurons
following pregnancy and pregnancy-like steroidal treatment in the rat. Brain
Res. Bull. 55, 737–745.
Khazipov, R., Tyzio, R., Ben-Ari, Y., 2008. Effects of oxytocin on GABA signalling in
the foetal brain during delivery. Prog. Brain Res. 170, 243–257.
Kim, P., Leckman, J.F., Mayes, L.C., Feldman, R., Wang, X., Swain, J.E., 2010. The
plasticity of human maternal brain: longitudinal changes in brainanatomy
during the early postpartum period. Behav. Neurosci. 124, 695–700.
Kim, P., Feldman, R., Mayes, L.C., Eicher, V., Thompson, N., Leckman, J.F., Swain, J.E.,
2011. Breastfeeding, brain activation to own infant cry, and maternal
sensitivity. J. Child Psychol. Psychiatry 52, 907–915.
Kingston, D., Tough, S., Whitfield, H., 2012. Prenatal and postpartum maternal
psychological distress and infant development: a systematic review.
Child Psychiat. Hum. Dev. 43, 683–714.
Kinsley, C.H., Amory-Meyer, E., 2011. Why the maternal brain? J. Neuroendocrinol.
23, 974–983.
Kinsley, C.H., Lambert, K.G., 2008. Reproduction-induced neuroplasticity: natural
behavioural and neuronal alterations associated with the production and care
of offspring. J. Neuroendocrinol. 20, 515–525.
Kinsley, C.H., Trainer, R., Stafisso-Sandoz, G., Quadros, P., Marcus, L.K., Hearon, C.,
Lambert, K.G., 2006. Motherhood and the hormones of pregnancy modify
concentrations of hippocampal neuronal dendritic spines. Horm. Behav. 49,
131–142.
Klampfl, S.M., Neumann, I.D., Bosch, O.J., 2013. Reduced brain corticotropin-
releasing factor receptor activation is required for adequate maternal care
and maternal aggression in lactating rats. Eur. J. Neurosci. 38, 2742–2750.
Kojima, S., Alberts, J.R., 2011. Oxytocin mediates the acquisition of filial, odor-
guided huddling for maternally-associated odor in preweanling rats. Horm.
Behav. 60, 549–558.
Kojima, S., Stewart, R.A., Demas, G.E., Alberts, J.R., 2012. Maternal contact
differentially modulates central and peripheral oxytocin in rat pups during a
brief regime of mother–pup interaction that induces a filial huddling
preference. J. Neuroendocrinol. 24, 831–840.
Koksma, J.J., Fritschy, J.M., Mack, V., Van Kesteren, R.E., Brussaard, A.B., 2005.
Differential GABAA receptor clustering determines GABA synapse plasticity in
rat oxytocin neurons around parturition and the onset of lactation. Mol. Cell.
Neurosci. 28, 128–140.
Krpan, K.M., Coombs, R., Zinga, D., Steiner, M., Fleming, A.S., 2005. Experiential and
hormonal correlates of maternal behavior in teen and adult mothers. Horm.
Behav. 47, 112–122.
Lagercrantz, H., Changeux, J.P., 2010. Basic consciousness of the newborn. Semin.
Perinatol. 34, 201–206.
Lagercrantz, H., Slotkin, T.A., 1986. The ‘‘stress’’ of being born. Sci. Am. 254, 100–
107.
Landgraf, R., Neumann, I., Pittman, Q.J., 1991. Septal and hippocampal release of
vasopressin and oxytocin during late pregnancy and parturition in the rat.
Neuroendocrinology 54, 378–383.
Landgraf, R., Neumann, I., Russell, J.A., Pittman, Q.J., 1992. Push-pull perfusion and
microdialysis studies of central oxytocin and vasopressin release in freely
moving rats during pregnancy, parturition, and lactation. Ann. N. Y. Acad. Sci.
652, 326–339.
Leng, G., Brown, C.H., Russell, J.A., 1999. Physiological pathways regulating the
activity of magnocellular neurosecretory cells. Prog. Neurobiol. 57, 625–655.
Leuner, B., Mirescu, C., Noiman, L., Gould, E., 2007. Maternal experience inhibits the
production of immature neurons in the hippocampus during the postpartum
period through elevations in adrenal steroids. Hippocampus 17, 434–442.
Levy, F., Keller, M., Poindron, P., 2004. Olfactory regulation of maternal behavior in
mammals. Horm. Behav. 46, 284–302.
Liao, J.B., Buhimschi, C.S., Norwitz, E.R., 2005. Normal labor: mechanism and
duration. Obstet. Gynecol. Clin. North Am. 32, 145–164.
Lincoln, D.W., Wakerley, J.B., 1974. Electrophysiological evidence for the activation
of supraoptic neurones during the release of oxytocin. J. Physiol. 242, 533–554.
Lipari, E.F., Lipari, D., Gerbino, A., Di Liberto, D., Bellafiore, M., Catalano, M.,
Valentino, B., 2001. The hypothalamic magnocellular neurosecretory system in
developing rats. Eur. J. Histochem. 45, 163–168.
Lord, C., Steiner, M., Soares, C.N., Carew, C.L., Hall, G.B., 2012. Stress response
in postpartum women with and without obsessive-compulsive symptoms: an
fMRI study. J. Psychiatry Neurosci. 37, 78–86.
Lumbiganon, P., Laopaiboon, M., Gulmezoglu, A.M., Souza, J.P., Taneepanichskul, S.,
Ruyan, P., World Health Organization Global Survey on Maternal and Perinatal
Health Research Group, 2010. Method of delivery and pregnancy outcomes in
asia: the WHO global survey on maternal and perinatal health 2007–08. Lancet
375, 490–499.
MacKay, D.F., Smith, G.C., Dobbie, R., Pell, J.P., 2010. Gestational age at delivery and
special educational need: Retrospective cohort study of 407,503 schoolchildren.
PLoS Med. 7, e1000289.
Mackeen, A.D., Fehnel, E., Berghella, V., Klein, T., 2014. Morphine sleep in pregnancy.
Am. J. Perinatol. 31, 085–090.
Maheu, M.E., Akbari, E.M., Fleming, A.S., 2009. Callosal oligodendrocyte number in
postpartum Sprague-Dawley rats. Brain Res. 1267, 18–24.
Malamitsi-Puchner, A., Protonotariou, E., Boutsikou, T., Makrakis, E., Sarandakou, A.,
Creatsas, G., 2005. The influence of the mode of delivery on circulating cytokine
concentrations in the perinatal period. Early Hum. Dev. 81, 387–392.
Malek, A., Blann, E., Mattison, D.R., 1996. Human placental transport of oxytocin. J.
Matern. Fetal Med. 5, 245–255.
Marco, E.M., Valero, M., de la Serna, O., Aisa, B., Borcel, E., Ramirez, M.J., Viveros,
M.P., 2013. Maternal deprivation effects on brain plasticity and recognition
memory in adolescent male and female rats. Neuropharmacology 68, 223–231.
Matthiesen, A.S., Ransjo-Arvidson, A.B., Nissen, E., Uvnas-Moberg, K., 2001.
Postpartum maternal oxytocin release by newborns: effects of infant hand
massage and sucking. Birth 28, 13–19.
Mazzuca, M., Minlebaev, M., Shakirzyanova, A., Tyzio, R., Taccola, G., Janackova, S.,
Gataullina, S., Ben-Ari, Y., Giniatullin, R., Khazipov, R., 2011. Newborn analgesia
mediated by oxytocin during delivery. Front. Cell. Neurosci. 5, 3.
McAnulty, P.A., Burns, L.M., 2004. Evaluation of the maternal and neonatal effects of
the oxytocin antagonist, atosiban, in a cross-fostering study in rats. Reprod.
Toxicol. 19, 65–70.
Meaney, M.J., Brake, W., Gratton, A., 2002. Environmental regulation of the
development of mesolimbic dopamine systems: a neurobiological mechanism
for vulnerability to drug abuse? Psychoneuroendocrinology 27, 127–138.
Mears, K., McAuliffe, F., Grimes, H., Morrison, J.J., 2004. Foetal cortisol in relation to
labour, intrapartum events and mode of delivery. J. Obstet. Gynaecol. 24, 129–
132.
Mehler, K., Wendrich, D., Kissgen, R., Roth, B., Oberthuer, A., Pillekamp, F., Kribs, A.,
2011. Mothers seeing their VLBW infants within 3 h after birth are more likely
470 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
to establish a secure attachment behavior: evidence of a sensitive period with
preterm infants? J. Perinatol. 31, 404–410.
Miller, S.S., Spear, N.E., 2008. Olfactory learning in the rat neonate soon after birth.
Dev. Psychobiol. 50, 554–565.
Miller, N.M., Fisk, N.M., Modi, N., Glover, V., 2005. Stress responses at birth:
determinants of cord arterial cortisol and links with cortisol response in
infancy. BJOG 112, 921–926.
Mogi, K., Nagasawa, M., Kikusui, T., 2011. Developmental consequences and
biological significance of mother–infant bonding. Prog.
Neuropsychopharmacol. Biol. Psychiatry 35, 1232–1241.
Moos, F., Poulain, D.A., Rodriguez, F., Guerne, Y., Vincent, J.D., Richard, P., 1989.
Release of oxytocin within the supraoptic nucleus during the milk ejection
reflex in rats. Exp. Brain Res. 76, 593–602.
Musser, E.D., Kaiser-Laurent, H., Ablow, J.C., 2012. The neural correlates of maternal
sensitivity: an fMRI study. Dev. Cogn. Neurosci. 2, 428–436.
Nagasawa, M., Okabe, S., Mogi, K., Kikusui, T., 2012. Oxytocin and mutual
communication in mother–infant bonding. Front. Hum. Neurosci. 6, 1–10.
National Center for Chronic Disease Prevention and Health Promotion, 2013.
Breastfeding Report Card United States/2013. Centers for Disease Control and
Prevention, Atlanta, GA.
Neumann, I., Russell, J.A., Wolff, B., Landgraf, R., 1991. Naloxone increases the
release of oxytocin, but not vasopressin, within limbic brain areas of conscious
parturient rats: a push-pull perfusion study. Neuroendocrinology 54, 545–551.
Neumann, I., Ludwig, M., Engelmann, M., Pittman, Q.J., Landgraf, R., 1993a.
Simultaneous microdialysis in blood and brain: oxytocin and vasopressin
release in response to central and peripheral osmotic stimulation and suckling
in the rat. Neuroendocrinol. 58, 637–645.
Neumann, I., Russell, J.A., Landgraf, R., 1993b. Oxytocin and vasopressin release
within the supraoptic and paraventricular nuclei of pregnant, parturient and
lactating rats: a microdialysis study. Neuroscience 53, 65–75.
Neumann, I.D., Johnstone, H.A., Hatzinger, M., Liebsch, G., Shipston, M., Russell, J.A.,
Landgraf, R., Douglas, A.J., 1998. Attenuated neuroendocrine responses to
emotional and physical stressors in pregnant rats involve adenohypophysial
changes. J. Physiol. 508 (Pt 1), 289–300.
Neumann, I.D., Torner, L., Wigger, A., 2000. Brain oxytocin: differential inhibition of
neuroendocrine stress responses and anxiety-related behaviour in virgin,
pregnant and lactating rats. Neuroscience 95, 567–575.
Neumann, I.D., Maloumby, R., Beiderbeck, D.I., Lukas, M., Landgraf, R., 2013.
Increased brain and plasma oxytocin after nasal and peripheral administration
in rats and mice. Psychoneuroendocrinology 38, 1985–1993.
NICHD Early Child Care Research Network, 1999. Child care and mother-child
interaction in the first 3 years of life. Dev. Psychol. 35, 1399–1413.
Nissen, E., Lilja, G., Widström, A.M., Uvnäs-Moberg, K., 1995. Elevation of oxytocin
levels early post partum in women. Acta Obstet. Gynecol. Scand. 74, 530–533.
Nissen, E., Uvnas-Moberg, K., Svensson, K., Stock, S., Widstrom, A.M., Winberg, J.,
1996. Different patterns of oxytocin, prolactin but not cortisol release during
breastfeeding in women delivered by caesarean section or by the vaginal route.
Early Hum. Dev. 45, 103–118.
Nissen, E., Gustavsson, P., Widstrom, A.M., Uvnas-Moberg, K., 1998. Oxytocin,
prolactin, milk production and their relationship with personality traits in
women after vaginal delivery or cesarean section. J. Psychosom. Obstet.
Gynaecol. 19, 49–58.
Nuttall, C., 2000. Caesarean section controversy. The caesarean culture of brazil. BMJ
320, 1074.
Nyqvist, K.H., Anderson, G.C., Bergman, N., Cattaneo, A., Charpak, N., Davanzo, R.,
Ewald, U., Ibe, O., Ludington-Hoe, S., Mendoza, S., Pallás-Allonso, C., Ruiz Peláez,
J.G., Sizun, J., Widström, A.M., 2010. Towards universal Kangaroo Mother Care:
recommendations and report from the First European conference and Seventh
International Workshop on Kangaroo Mother Care. Acta Paediatr. 99, 820–826.
Oatridge, A., Holdcroft, A., Saeed, N., Hajnal, J.V., Puri, B.K., Fusi, L., Bydder, G.M.,
2002. Change in brain size during and after pregnancy: study in healthy women
and women with preeclampsia. Am. J. Neuroradiol. 23, 19–26.
Odent, M., 2009. The instincts of motherhood: bringing joy back into newborn care.
Early Hum. Dev. 85, 697–700.
Ohana, E., Mazor, M., Chaim, W., Levy, J., Sharoni, Y., Leiberman, J.R., Glezerman, M.,
1996. Maternal plasma and amniotic fluid cortisol and progesterone
concentrations between women with and without term labor. A comparison.
J. Reprod. Med. 41, 80–86.
Olza Fernandez, I., Marin Gabriel, M., Malalana Martinez, A., Fernandez-Canadas
Morillo, A., Lopez Sanchez, F., Costarelli, V., 2012. Newborn feeding behaviour
depressed by intrapartum oxytocin: a pilot study. Acta Paediatr. 101, 749–754.
Olza Fernandez, I., Marin Gabriel, M.A., Garcia Murillo, L., Malalana Martinez, A.M.,
Costarelli, V., Millan Santos, I., 2013. Mode of delivery may influence neonatal
responsiveness to maternal separation. Early Hum. Dev. 89, 339–342.
Ounsted, M.K., Boyd, P.A., Hendrick, A.M., Mutch, L.M., Simons, C.D., Good, F.J.,
1978a. Induction of labour by different methods in primiparous women. II.
neuro-behavioural status of the infants. Early Hum. Dev. 2, 241–253.
Ounsted, M.K., Hendrick, A.M., Mutch, L.M., Calder, A.A., Good, F.J., 1978b. Induction
of labour by different methods in primiparous women. I some perinatal and
postnatal problems. Early Hum. Dev. 2, 227–239.
Papatsonis, D.N., Flenady, V., Liley, H., 2013. Maintenance therapy with oxytocin
antagonists for inhibiting preterm birth after threatened preterm labour.
Cochrane Database Syst. Rev. 10, CD005938.
Pawluski, J.L., Galea, L.A., 2006. Hippocampal morphology is differentially affected
by reproductive experience in the mother. J. Neurobiol. 66, 71–81.
Pawluski, J.L., Galea, L.A., 2007. Reproductive experience alters hippocampal
neurogenesis during the postpartum period in the dam. Neuroscience 149,
53–67.
Pawluski, J.L., Brummelte, S., Barha, C.K., Crozier, T.M., Galea, L.A., 2009. Effects of
steroid hormones on neurogenesis in the hippocampus of the adult female
rodent during the estrous cycle, pregnancy, lactation and aging. Front.
Neuroendocrinol. 30, 343–357.
Pawluski, J.L., van den Hove, D.L., Rayen, I., Prickaerts, J., Steinbusch, H.W., 2011.
Stress and the pregnant female: impact on hippocampal cell proliferation, but
not affective-like behaviors. Horm. Behav. 59, 572–580.
Pawluski, J.L., Charlier, T.D., Fillet, M., Houbart, V., Crispin, H.T., Steinbusch, H.W.,
van den Hove, D.L., 2012. Chronic fluoxetine treatment and maternal adversity
differentially alter neurobehavioral outcomes in the rat dam. Behav. Brain Res.
228, 159–168.
Pierrehumbert, B., Nicole, A., Muller-Nix, C., Forcada-Guex, M., Ansermet, F., 2003.
Parental post-traumatic reactions after premature birth: implications for
sleeping and eating problems in the infant. Arch. Dis. Child. Fetal Neonatal
Ed. 88, F400–F404.
Porter, R.H., Winberg, J., 1999. Unique salience of maternal breast odors for
newborn infants. Neurosci. Biobehav. Rev. 23, 439–449.
Poulain, D.A., Wakerley, J.B., 1982. Electrophysiology of hypothalamic
magnocellular neurones secreting oxytocin and vasopressin. Neuroscience 7,
773–808.
Preis, K., Bidzan, M., Swiatkowska-Freund, M., Peplinska, A., 2012. Long-term
follow-up for organic dysfunction in breech – presenting children. Med. Sci.
Monit. 18, CR741–CR746.
Prevost, M., Zelkowitz, P., Tulandi, T., Hayton, B., Feeley, N., Carter, C.S., Joseph, L.,
Pournajafi-Nazarloo, H., Yong Ping, E., Abenhaim, H., Gold, I., 2014. Oxytocin in
pregnancy and the postpartum: relations to labor and its management. Front.
Public Health 2, 1.
Puolakka, J., Kauppila, A., Tuimala, R., Jouppila, R., Vuori, J., 1983. The effect
of parturition on umbilical blood plasma levels of norepinephrine. Obstet.
Gynecol. 61, 19–21.
Raine, A., Brennan, P., Mednick, S.A., 1994. Birth complications combined with early
maternal rejection at age 1 year predispose to violent crime at age 18 years.
Arch. Gen. Psychiatry 51, 984–988.
Raine, A., Brennan, P., Mednick, S.A., 1997. Interaction between birth complications
and early maternal rejection in predisposing individuals to adult violence:
specificity to serious, early-onset violence. Am. J. Psychiatry 154, 1265–1271.
Rasia-Filho, A.A., Fabian, C., Rigoti, K.M., Achaval, M., 2004. Influence of sex, estrous
cycle and motherhood on dendritic spine density in the rat medial amygdala
revealed by the Golgi method. Neuroscience 126, 839–847.
Rauh, C., Beetz, A., Burger, P., Engel, A., Haberle, L., Fasching, P.A., Faschingbauer, F.,
2012. Delivery mode and the course of pre- and postpartum depression. Arch.
Gynecol. Obstet. 286, 1407–1412.
Roberts, J., Hanson, L., 2007. Best practices in second stage labor care: maternal
bearing down and positioning. J. Midwifery Womens Health 52, 238–245.
Ronca, A.E., Alberts, J.R., 1995. Simulated uterine contractions facilitate fetal and
newborn respiratory behavior in rats. Physiol. Behav. 58, 1035–1041.
Ronca, A.E., Abel, R.A., Ronan, P.J., Renner, K.J., Alberts, J.R., 2006. Effects of labor
contractions on catecholamine release and breathing frequency in newborn
rats. Behav. Neurosci. 120, 1308–1314.
Rupp, H.A., James, T.W., Ketterson, E.D., Sengelaub, D.R., Ditzen, B., Heiman, J.R.,
2014. Amygdala response to negative images in postpartum vs
nulliparouswomen and intranasal oxytocin. Soc. Cogn. Affect. Neurosci. 9, 48–
54.
Russell, J.A., Leng, G., Douglas, A.J., 2003. The magnocellular oxytocin system, the
fount of maternity: adaptations in pregnancy. Front. Neuroendocrinol. 24, 27–
61.
Ryding, E.L., Wijma, B., Wijma, K., 1997. Posttraumatic stress reactions after
emergency cesarean section. Acta Obstet. Gynecol. Scand. 76, 856–861.
Ryding, E.L., Wijma, K., Wijma, B., 1998. Psychological impact of emergency
cesarean section in comparison with elective cesarean section, instrumental
and normal vaginal delivery. J. Psychosom. Obstet. Gynaecol. 19, 135–144.
Salmaso, N., Cossette, M.P., Woodside, B., 2011. Pregnancy and maternal behavior
induce changes in glia, glutamate and its metabolism within the cingulate
cortex. PLoS ONE, e23529.
Schlinzig, T., Johansson, S., Gunnar, A., Ekstrom, T.J., Norman, M., 2009. Epigenetic
modulation at birth altered DNA-methylation in white blood cells after
cesarean section. Acta Paediatr. 98, 1096–1099.
Schubert, F., George, J.M., Rao, M.B., 1981. Vasopressin and oxytocin content of
human fetal brain at different stages of gestation. Brain Res. 213, 111–117.
Shaw, R.J., Bernard, R.S., Deblois, T., Ikuta, L.M., Ginzburg, K., Koopman, C., 2009. The
relationship between acute stress disorder and posttraumatic stress disorder in
the neonatal intensive care unit. Psychosomatics 50, 131–137.
Simon-Areces, J., Dietrich, M.O., Hermes, G., Garcia-Segura, L.M., Arevalo, M.,
Horvath, T.L., 2012. Ucp2 induced by natural birth regulates neuronal
differentiation of the hippocampus and related adult behavior. PLoS ONE 7,
e42911.
Slattery, D.A., Neumann, I.D., 2008. No stress please! mechanisms of stress
hyporesponsiveness of the maternal brain. J. Physiol. 586, 377–385.
Smith, J., Plaat, F., Fisk, N.M., 2008. The natural caesarean: a woman-centred
technique. BJOG 115, 1037–1042.
Soderquist, J., Wijma, K., Wijma, B., 2002. Traumatic stress after childbirth: the role
of obstetric variables. J. Psychosom. Obstet. Gynaecol. 23, 31–39.
I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472 471
Souza, J. P., Gulmezoglu, A., Lumbiganon, P., Laopaiboon, M., Carroli, G., Fawole, B.,
WHO Global Survey on Maternal and Perinatal Health Research Group., 2010.
Caesarean section without medical indications is associated with an increased
risk of adverse short-term maternal outcomes: The 2004–2008 WHO global
survey on maternal and perinatal health. BMC Med. 8, 71.
Stallings, J., Fleming, A.S., Corter, C., Worthman, C., Steiner, M., 2001. The effects of
infant cries and odors on sympathy, cortisol and autonomic responses in new
mothers and nonpostpartum women. Parent. Sci. Pract. 1, 71–100.
Stern, J.M., Goldman, L., Levine, S., 1973. Pituitary-adrenal responsiveness
during lactation in rats. Neuroendocrinol. 12, 179–191.
Stern, J.E., Hestrin, S., Armstrong, W.E., 2000. Enhanced neurotransmitter release at
glutamatergic synapses on oxytocin neurones during lactation in the rat. J.
Physiol. 526, 109–114.
Strathearn, L., 2011. Maternal neglect: oxytocin, dopamine and the neurobiology of
attachment. J. Neuroendocrinol. 23, 1054–1065.
Strathearn, L., Fonagy, P., Amico, J., Montague, P.R., 2009a. Adult attachment
predicts maternal brain and oxytocin response to infant cues.
Neuropsychopharmacology 34, 2655–2666.
Strathearn, L., Mamun, A.A., Najman, J.M., O’Callaghan, M.J., 2009b. Does
breastfeeding protect against substantiated child abuse and neglect? A 15-
year cohort study. Pediatrics 123, 483–493.
Sullivan, R.M., McGaugh, J.L., Leon, M., 1991. Norepinephrine-induced plasticity and
one-trial olfactory learning in neonatal rats. Brain Res. Dev. Brain Res. 60, 219–
228.
Swain, J.E., Tasgin, E., Mayes, L.C., Feldman, R., Constable, R.T., Leckman, J.F., 2008.
Maternal brain response to own baby-cry is affected by cesarean section
delivery. J. Child Psychol. Psychiatry 49, 1042–1052.
Szabo, A.L., 2013. Review article: intrapartum neuraxial analgesia and breastfeeding
outcomes: limitations of current knowledge. Anesth. Analg. 116, 399–405.
Takagi, T., Tanizawa, O., Otsuki, Y., Haruta, M., Yamaji, K., 1985. Oxytocin in the
cerebrospinal fluid and plasma of pregnant and non-pregnant subjects. Horm.
Metab. Res. 17, 308–310.
Takeda, S., Kiwabara, Y., Mizuno, M., 1985. Effects of pregnancy and labor on
oxytocin levels in human plasma and cerebrospinal fluid. Endocrinol. Jpn. 32,
875–880.
Tasker, J.G., Di, S., Boudaba, C., 2002. Functional synaptic plasticity in hypothalamic
magnocellular neurons. Prog. Brain Res. 139, 113–119.
Theodosis, D.T., Poulain, D.A., 2001. Maternity leads to morphological synaptic
plasticity in the oxytocin system. Prog. Brain Res. 133, 49–58.
Theodosis, D.T., Trailin, A., Poulain, D.A., 2006. Remodeling of astrocytes, a
prerequisite for synapse turnover in the adult brain? insights from the
oxytocin system of the hypothalamus. Am. J. Physiol. Regul. Integr. Comp.
Physiol. 290, R1175–R1182.
Torner, L., Neumann, I.D., 2002. The brain prolactin system: involvement in stress
response adaptations in lactation. Stress 5, 249–257.
Tyzio, R., Cossart, R., Khalilov, I., Minlebaev, M., Hübner, C.A., Represa, A., Ben-Ari, Y.,
Khazipov, R., 2006. Maternal oxytocin triggers a transient inhibitory switch in
GABA signaling in the fetal brain during delivery. Science 314, 1788–1792.
Tyzio, R., Cossart, R., Khalilov, I., Represa, A., Ben-Ari, Y., Khazipov, R., 2007.
Response to comment on ‘‘Maternal oxytocin triggers a transient inhibitory
switch in GABA signaling in the fetal brain during delivery’’. Science 317, 197.
Tyzio, R., Nardou, R., Ferrari, D.C., Tsintsadze, T., Shahrokhi, A., Eftekhari, S., Khalilov,
I., Tsintsadze, V., Brouchoud, C., Chazal, G., Lemonnier, E., Lozovaya, N.,
Burnashev, N., Ben-Ari, Y., 2014. Oxytocin-mediated GABA inhibition during
delivery attenuates autism pathogenesis in rodent offspring. Science 343, 675–
679.
Uvnäs-Moberg, K., 1996. Neuroendocrinology of the mother–child interaction.
Trends Endocrinol. Metab. 7, 126–131.
Uvnäs-Moberg, K., Widström, A.M., Werner, S., Matthiesen, A.S., Winberg, J., 1990.
Oxytocin and prolactin levels in breast-feeding women. Correlation with milk
yield and duration of breast-feeding. Acta Obstet. Gynecol. Scand. 69, 301–306.
Valenzuela, G.J., Craig, J., Bernhardt, M.D., Holland, M.L., 1995. Placental passage of
the oxytocin antagonist atosiban. Am. J. Obstet. Gynecol. 172, 1304–1306.
Varendi, H., Porter, R.H., 2001. Breast odour as the only maternal stimulus elicits
crawling towards the odour source. Acta Paediatr. 90, 372–375.
Varendi, H., Porter, R.H., Winberg, J., 2002. The effect of labor on olfactory exposure
learning within the first postnatal hour. Behav. Neurosci. 116, 206–211.
Veenema, A.H., Neumann, I.D., 2008. Central vasopressin and oxytocin release:
regulation of complex social behaviours. Prog. Brain Res. 170, 261–276.
Velandia, M., Matthisen, A.S., Uvnas-Moberg, K., Nissen, E., 2010. Onset of vocal
interaction between parents and newborns in skin-to-skin contact immediately
after elective cesarean section. Birth 37, 192–201.
Virani, S., Dolinoy, D.C., Halubai, S., Jones, T.R., Domino, S.E., Rozek, L.S., Nahar, M.S.,
Padmanabhan, V., 2012. Delivery type not associated with global methylation at
birth. Clin. Epigenetics 4, 8.
Vogl, S.E., Worda, C., Egarter, C., Bieglmayer, C., Szekeres, T., Huber, J., Husslein, P.,
2006. Mode of delivery is associated with maternal and fetal endocrine stress
response. BJOG 113, 441–445.
Wahl, R.U., 2004. Could oxytocin administration during labor contribute to autism
and related behavioral disorders? – a look at the literature. Med. Hypotheses 63,
456–460.
Walker, C.D., Toufexis, D.J., Burlet, A., 2001. Hypothalamic and limbic expression of
CRF and vasopressin during lactation: implications for the control of ACTH
secretion and stress hyporesponsiveness. Prog. Brain Res. 133, 99–110.
Wankaew, N., Jirapradittha, J., Kiatchoosakun, P., 2013. Neonatal morbidity and
mortality for repeated cesarean section vs. normal vaginal delivery to
uncomplicated term pregnancies at Srinagarind Hospital. J. Med. Assoc. Thai.
96, 654–660.
Wellmann, S., Buhrer, C., 2012. Who plays the strings in newborn analgesia at birth,
vasopressin or oxytocin? Front. Neurosci. 6, 78.
Wellmann, S., Benzing, J., Cippa, G., Admaty, D., Creutzfeldt, R., Mieth, R.A., Bucher,
H.U., 2010. High copeptin concentrations in umbilical cord blood after vaginal
delivery and birth acidosis. J. Clin. Endocrinol. Metab. 95, 5091–5096.
White-Traut, R., Watanabe, K., Pournajafi-Nazarloo, H., Schwertz, D., Bell, A., Carter,
C.S., 2009. Detection of salivary oxytocin levels in lactating women. Dev.
Psychobiol. 51, 367–373.
World Health Organization, 2003. Global Strategy for Infant and Young Child
Feeding. World Health Organization, Geneva.
Widstrom, A.M., Ransjo-Arvidson, A.B., Christensson, K., Matthiesen, A.S., Winberg,
J., Uvnas-Moberg, K., 1987. Gastric suction in healthy newborn infants. Effects
on circulation and developing feeding behaviour. Acta Paediatr. Scand. 76, 566–
572.
Wieczorek, P.M., Guest, S., Balki, M., Shah, V., Carvalho, J.C., 2010. Breastfeeding
success rate after vaginal delivery can be high despite the use of epidural
fentanyl: an observational cohort study. Int. J. Obstet. Anesth. 19, 273–277.
Wiesenfeld, A.R., Malatesta, C.Z., Whitman, P.B., Granrose, C., Uili, R., 1985.
Psychophysiological response of breast- and bottle-feeding mothers to their
infants’ signals. Psychophysiology 22, 79–86.
Wigger, A., Lörscher, P., Oehler, I., Keck, M.E., Naruo, T., Neumann, I.D., 1999.
Nonresponsiveness of the rat hypothalamo-pituitary-adrenocortical axis to
parturition-related events: inhibitory action of endogenous opioids.
Endocrinology 140, 2843–2849.
Wijma, K., Soderquist, J., Wijma, B., 1997. Posttraumatic stress disorder after
childbirth: a cross sectional study. J. Anxiety Disor. 11, 587–597.
Wiklund, I., Norman, M., Uvnas-Moberg, K., Ransjo-Arvidson, A.B., Andolf, E., 2009.
Epidural analgesia: breast-feeding success and related factors. Midwifery 25,
e31–e38.
Willcox, D.L., Yovich, J.L., McColm, S.C., Phillips, J.M., 1985. Progesterone, cortisol
and oestradiol-17 beta in the initiation of humanparturition: partitioning
between free and bound hormone in plasma. Br. J. Obstet. Gynaecol. 92, 65–71.
Wilson, M.J., MacArthur, C., Cooper, G.M., Bick, D., Moore, P.A., Shennan, A., COMET
Study Group UK, 2010. Epidural analgesia and breastfeeding: a randomised
controlled trial of epidural techniques with and without fentanyl and a non-
epidural comparison group. Anaesthesia 65, 145–153.
Winberg, J., 2005. Mother and newborn baby: mutual regulation of physiology and
behavior–a selective review. Dev. Psychobiol. 47, 217–229.
Windle, R.J., Gamble, L.E., Kershaw, Y.M., Wood, S.A., Lightman, S.L., Ingram, C.D.,
2006. Gonadal steroid modulation of stress-induced hypothalamo-pituitary-
adrenal activity and anxiety behavior: role of central oxytocin. Endocrinology
147, 2423–2431.
Workman, J.L., Barha, C.K., Galea, L.A., 2012. Endocrine substrates of cognitive and
affective changes during pregnancy and postpartum. Behav. Neurosci. 126, 54–
72.
Yektaei-Karin, E., Moshfegh, A., Lundahl, J., Berggren, V., Hansson, L.O., Marchini, G.,
2007. The stress of birth enhances in vitro spontaneous and IL-8-induced
neutrophil chemotaxis in the human newborn. Pediatr. Allergy Immunol. 18,
643–651.
Yeo, J.A.G., Keverne, E.B., 1986. The importance of vaginal–cervical stimulation for
maternal behaviour in the rat. Physiol. Behav. 37, 23–26.
Young, D., 2011. ‘‘Gentle cesareans’’: better in some respects, but fewer cesareans
are better still. Birth 38, 183–184.
Zanardo, V., Savona, V., Cavallin, F., D’Antona, D., Giustardi, A., Trevisanuto, D., 2012.
Impaired lactation performance following elective delivery at term: role of
maternal levels of cortisol and prolactin. J. Matern. Fetal Neonat. Med. 25,
1595–1598.
Zanardo, V., Pigozzo, A., Wainer, G., Marchesoni, D., Gasparoni, A., Di Fabio, S.,
Trevisanuto, D., 2013. Early lactation failure and formula adoption after elective
caesarean delivery: cohort study. Arch. Dis. Child. Fetal Neonatal Ed. 98, F37–
F41.
472 I. Olza-Fernández et al. / Frontiers in Neuroendocrinology 35 (2014) 459–472
... Por otra parte, actualmente la atención clínica de los partos considera una serie de intervenciones biotecnológicas protocolizadas 4 , lo cual ha traído consecuencias, como el aumento en la medicalización de los partos de embarazos normales y en la tasa de cesáreas, sobre todo en los países de ingresos medios y altos 5 . Pero el parto, más allá de ser un evento mecánico, es un evento neuroendocrino e involucra diversas neurohormonas, y entre las más reconocidas se encuentran la oxitocina y el cortisol, y también otras que podrían verse involucradas en el neurodesarrollo del neonato, como las hormonas tiroideas [6][7][8] . ...
... La literatura reconoce en el parto vaginal beneficios neurológicos e inmunológicos para la salud del recién nacido. A su vez, el parto es considerado un evento crítico, puesto que tanto las prácticas clínicas como la vía de parto pueden modificar su proceso fisiológico, lo que podría implicar efectos epigenéticos para la adaptación materna o neonatal 7,[9][10][11] . En este contexto, medir las hormonas en plasma del cordón umbilical en el parto, tales como la OT (relacionada con el apego y la socialización) 12 , el cortisol (relacionado con el estrés y la adaptación al nacimiento) 13 y las hormonas tiroideas (asociadas al neurodesarrollo) 14,15 , resultaría un aporte significativo a la práctica obstétrica, orientado a disminuir el efecto cascada de las intervenciones clínicas 16 . ...
Article
Full-text available
Introducción: La placenta sintetiza y secreta varias hormonas que permiten la regulación del embarazo, el trabajo de parto y la adaptación metabólica materno-fetal. Su comportamiento asociado al tipo de parto puede dar información relevante sobre efectos epigenéticos. Objetivo: Describir el tipo de parto con los niveles de oxitocina, cortisol y hormonas tiroideas en plasma de cordón umbilical al nacer. Método: A 50 mujeres con embarazos principalmente normales se les cuantificaron los niveles neurohormonales en plasma de cordón umbilical, obtenido inmediatamente tras el periodo expulsivo. Los resultados se incorporaron a la base de datos clínicos de cada participante y se analizaron con Stata v.14.0. El protocolo fue aprobado por el comité de ética. Resultados: Hubo 33 partos vaginales (12 espontáneos, 13 acelerados y 8 inducidos) y 17 cesáreas (7 electivas y 10 de urgencia). Se observaron mayores niveles de cortisol en los partos vaginales acelerados; las cesáreas tuvieron menores niveles de cortisol y hormona estimulante de la tiroides. Las intervenciones clínicas, con altos o bajos niveles hormonales, están en directa relación con el tipo de parto. Conclusiones: El cortisol y la hormona estimulante de la tiroides medidos en plasma de cordón umbilical variaron según el tipo de parto. Esto es una primera cuantificación de hormonas en plasma de cordón umbilical y su posible regulación placentaria a propósito del tipo de parto.
... In case it is the woman's will, and she presents favorable clinical conditions, the baby should be placed in direct contact-skin to skin-with the mother, naked and without physical barriers between them (e.g., fields and compresses) to promote oxytocin production. Studies show that the effect of this contact from the senses causes serum increases in oxytocin in both woman and neonate, with advantages observed for both (Uvnas-Moberg, 1998;Olza-Fernández et al., 2014). ...
... Skin-to-skin contact, if performed uninterruptedly, promotes a faster physiological uterine involution and a reduction in the volume of postpartum bleeding (Uvnas-Moberg, 1998;Olza-Fernández, et al, 2014;Salam, et al., 2014;Saxton et al., 2014;Abdelmenem et al., 2019), as shown in the results of the included studies. However, as discussed, in most studies, there is no description of contact time and whether it occurred without interruptions by the team. ...
Article
Full-text available
Introduction Literature supports numerous benefits of skin-to-skin contact for neonatal adaptation to extrauterine life and bonding/attachment, but few studies explore the effects of skin-to-skin contact on maternal outcomes. This review aims to map the evidence on skin-to-skin contact in the third stage of labor for postpartum hemorrhage prevention. Methods Scoping review, which covered stages recommended by the Institute Joanna Briggs, including studies from the PubMed, EMBASE, CINAHL, LILACS, Web of Science, and Scopus databases, using the descriptors “Postpartum hemorrhage”, “Labor stages, third”, “Prevention” and “Kangaroo care/Skin-to-skin”. Results 100 publications on the subject found, 13 articles met the inclusion criteria, with 10,169 dyads were assessed in all studies. Publications from 2008 to 2021 were mostly written in English and designed as a randomized controlled trial. Skin-to-skin contact was effective and significant in: reducing the duration of the third stage of labor; placenta delivery; uterine contractility and physiological involution; absence of atony, decreasing blood loss with lower rates of erythrocyte and hemoglobin drop; reducing the need for synthetic oxytocin and/or ergometrine to control bleeding; and reducing changing pads per period and length of stay. Discussion Skin-to-skin contact was considered an effective, low-cost, and safe strategy, with positive effects already established in the literature for infants and extremely favorable results in postpartum hemorrhage prevention cases, being highly recommended in assistance for the dyad. Open Science Framework Registry (https://osf.io/n3685).
... Hormonal levels in both mother and baby reach higher levels than at any other time in life. Actions as simple as separation from the mother after childbirth can impact the beginning of attachment, bonding, breastfeeding, and the neurodevelopment of the newborn [64]. ...
Chapter
Full-text available
All the tissues of our body are formed by cells, but there are at least 1–10 times more microbes than cells in it, which coexist in balance and help us maintain our health. Most of them are found in the intestine; through their interaction with the food, they produce a great variety of functions related to nutrition and the functioning of the nervous, immune, and cardiovascular systems through their influence on gene programming, among others. It is now known that there are periods in life that are more sensitive and have greater long-term impacts that depend on the proper functioning of this large number of microbes in the human body. Pregnancy, birth, and the first 2 years of the life of a human are crucial stages that can predispose a person to health or disease, and in this chapter, we will be able to delve into the role played by cesarean section as a way of birth in this regard.
... Maximum levels of endogenous oxytocin are achieved within 1 h of delivery in both maternal and infant brains. Maternal oxytocin release increases via skin-to-skin contact with the infant 6,7 . This hormone promotes uterine restoration in postpartum mothers and is an essential hormone released during breastfeeding that causes the breast milk ejection reflex 8 . ...
Article
Full-text available
This study aimed to examine the effect of intrapartum oxytocin administration on neonatal sucking behavior and breastfeeding. A total of 64 pairs (29 in the group treated with intrapartum oxytocin and 35 in the control group) of normal infants within 24–48 h of birth and their mothers were recruited. Sucking ability was evaluated by measuring Non-Nutritive Sucking (NNS) for 5 min. Data on the rate of exclusive breastfeeding at 1 month postpartum were collected. In the adjusted multiple regression models, intrapartum oxytocin exposure was significantly associated with fewer total NNS bursts (95% confidence interval (CI), −7.02 to −0.22), longer pause times (95% CI, 1.33 to 10.21), and greater pause-time variability (95% CI, 3.63 to 63.92). Effects estimated using structural equation modeling revealed that intrapartum oxytocin exposure had a significant negative and direct effect on the practice of exclusive breastfeeding 1 month postpartum (β = −0.238, p = 0.047). However, no NNS-mediated indirect effects were observed. This report demonstrates that infants born to mothers who receive intrapartum oxytocin may have impaired sucking ability for at least the first 48 h after birth, and breastfeeding support should be provided.
... To begin with, breastfeeding involves increased production of oxytocin that also affects how quickly cortisol levels drop during breastfeeding. Higher levels of circulating oxytocin are associated with greater activation of the brain's mesocorticolimbic reward system (9). Therefore, a decrease in oxytocin levels might contribute to the unpleasant symptoms experienced in D-MER. ...
Article
Full-text available
Given its endless benefits, breastfeeding is widely acknowledged as the optimal choice for both maternal and infant health. Nevertheless, breastfeeding mothers often encounter various challenges that may hinder their ability to fully embrace this experience. This report delves into a compelling case of Dysphoric Milk Ejection Reflex (D-MER), a largely underexamined mental health issue among lactating mothers. D-MER is characterized by intense aversion right before milk let down, which can significantly impede a mother’s willingness to breastfeed. The primary aim of this case report is to provide a comprehensive psychological understanding of D-MER, emphasizing aspects of attachment, the transition into motherhood, and the sociocultural sexualization of the female body. We also offer an overview of the psychotherapeutic journey, highlighting key insights and progress achieved over a span of six months. Therapy adopted an integrative approach combining narrative techniques and skills training such as mindfulness to facilitate a comprehensive therapeutic experience. This case underscores the psychological dimensions of the breastfeeding experience, complementing the well-established biochemical and physiological aspects of D-MER. It also emphasizes the need for further research into the psychological facets of both successful and less successful breastfeeding experiences.
... During the first hour after birth, both the mother and newborn experience a sensitive phase that is programmed by physiological factors, particularly in cases of vaginal delivery [1]. This period benefits from high levels of oxytocin in the mother and exceptionally elevated catecholamines in the infant to facilitate this state [2]. Early contact also plays an impactful role in establishing lasting microbiomes over extended periods [3]. ...
Article
Full-text available
This research project examines the long-term effects of maternal-neonatal bonding during a mother's "sensitive period." The review explores how early contact between a mother and her newborn can affect their psychosocial and emotional well-being in the future. Within an hour after birth, oxytocin levels increase for mothers, while catecholamine surges enhance neonates' memory retention to encourage immediate skin-to-skin contact (SSC), which promotes breastfeeding with benefits, such as quicker placenta expulsion, less bleeding, and lower stress. As per sources to date, there is no systematic review on this subject; however, numerous studies exist regarding short-term outcomes, exclusive breastfeeding, and childhood problems. The exploration involves rigorous searches of academic databases following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for transparency and reproducibility by using the Population, Intervention, Comparison, and Outcome (PICO) framework. Of the 516 initially identified articles, only five were relevant based on refined selection criteria, making it clear from the analysis that sensitive-period bonding produces long-term impacts in infants. Few studies are available, particularly in recent years; thus, more research is required in this area.
... Research on rats linked the pressure experienced during vaginal birth to effective neurophysiological adaption (respiration and suckling) in extrauterine life [13]. In humans, the pressure causes activation of the sympathoadrenal axis, with release of noradrenaline, vasopressin, and other hormones [14]. These hormones have various effects during the perinatal period on the mother (facilitate maternal behaviours and stimulate milk production) and on the infant (aid lung maturation, infant alert state, and olfactory recognition of the nipple). ...
Article
Full-text available
Background Research has shown that pressure in the vaginal birth process aids the infant's neurophysiological adaption to extrauterine life, including their ability to regulate their responses to diverse sensory stimuli. As pressure is absent in elective caesarean section births, we hypothesised that these children may be at higher risk for developing sensory hyperreactivity (SHR), a sensory modulation difficulty which negatively impacts on the child's engagement in their occupations. This paper reports on a study which investigated associations between birth method and SHR. Method In this cross-sectional study, 91 children aged three and four years from various language, cultural, and socioeconomic groups were recruited and categorised based on birth method (caesarean section or vaginal birth). Caregivers of each child completed the Short Sensory Profile-2 and a demographic questionnaire. The prevalence of SHR between the birth method groups was compared. Results The prevalence of SHR was greater in the vaginal birth group (29%) than the caesarean section group (9%). Mothers in the vaginal birth group were younger (p ≤ 0.001), of lower-income level (p = 0.003), and more likely to be single (p = 0.037). During data collection with the vaginal birth group, comprehensibility of certain items in the Short Sensory Profile-2 proved challenging for many caregivers. Conclusion The statistically significant higher prevalence of SHR in the vaginal birth group supported a null hypothesis. However, the results are questioned due to the challenges related to data collection. Given this, the study supports the need for further investigation of how sociocultural and socioeconomic factors influence the assessment of SHR in the South African context.
... Additionally, parents, especially mothers, who are at a higher risk of having children with ASD, may benefit from specific education and guidance regarding recognizing and responding to abnormal behaviors or responses during skin-to-skin contact through breastfeeding. Enhancing their awareness and understanding of these interactions can contribute to improved mother-infant emotional bonding and attachment, which are vital aspects of breastfeeding [63]. By addressing these considerations and providing targeted support to parents of ASD children and high-risk parents, we can strive towards optimizing breastfeeding practices and ensuring that the specific needs of these individuals and families are met effectively. ...
Article
Full-text available
Objective To investigate the relationship between infant feeding practices and autism spectrum disorder (ASD) among children aged 2–5 years in the United States (US). Methods Data from the 2016–2020 National Survey of Children’s Health, a nationally representative cross-sectional survey, were utilized for this study. Questionnaires were administered to parents of children aged 2–5 years to gather information on ASD diagnosis, infant feeding practices, and demographic factors (e.g., child sex, ethnic group, and maternal age at birth). Logistic regression with sample weights was employed to assess the association between infant feeding practices and ASD, while controlling for demographic variables. Polynomial regression models were used to examine trends in exclusive breastfeeding and ever breastfeeding rates among children with and without ASD. Results A total of 35,050 children aged 2–5 years were analyzed, including 616 diagnosed with ASD, after excluding participants with missing information on breastfeeding and ASD diagnosis. Of these children with ASD, 76.6% (n = 472) had a breastfeeding history, with 67.5% (n = 416) engaged in partial breastfeeding and 9.1% (n = 56) exclusively breastfed. Adjusted odds ratios for each additional month of breastfeeding compared to never being breastfed were 0.98 (95% CI, 0.96–1.01). The adjusted odds ratios for breastfeeding durations of > 0–6 months, > 6–12 months, > 12–24 months, and > 24 months were 0.81 (95% CI, 0.50–1.31), 0.65 (95% CI, 0.36–1.18), 0.81 (95% CI, 0.44–1.49), and 0.48 (95% CI, 0.23–1.01), respectively. Compared to children who were never breastfed, the adjusted odds ratio for children who were ever breastfed was 0.74 (95% CI, 0.47–1.18). Among children with ASD, the proportion of ever breastfeeding declined from 82.0% in 2017 to 64.3% in 2020, while exclusive breastfeeding decreased from 12.0% in 2016 to 4.2% in 2020. Conclusions and relevance Although no significant association was found between infant feeding practices and ASD among US children aged 2–5 years, the rates of breastfeeding, particularly exclusive breastfeeding, were suboptimal among children with ASD. This highlights the need for specific policies and practices to promote and support breastfeeding among parents of children with ASD or those at high risk of having a child with ASD.
Preprint
Full-text available
This study aimed to examine the effect of intrapartum oxytocin administration on neonatal sucking behavior and breastfeeding. A total of 64 pairs (29 in the group treated with intrapartum oxytocin and 35 in the control group) of normal infants within 24–48 h of birth and their mothers were recruited. Sucking ability was evaluated by measuring Non-Nutritive Sucking (NNS) for 5 min. Data on the rate of exclusive breastfeeding at 1 month postpartum were collected. In the adjusted multiple regression models, intrapartum oxytocin exposure was significantly associated with fewer total NNS bursts (95% confidence interval (CI), -6.811 to -0.311), longer pause times (95% CI, 0.320 to 9.223), and greater pause-time variability (95% CI, 4.453 to 62.738). Effects estimated using structural equation modeling revealed that intrapartum oxytocin exposure had a significant negative and direct effect on the practice of exclusive breastfeeding 1 month postpartum (β=-.238, p = 0.047). However, no NNS-mediated indirect effects were observed. This report demonstrates that infants born to mothers who receive intrapartum oxytocin may have impaired sucking ability for at least the first 48 h after birth, and breastfeeding support should be provided.
Article
The amount of stress experienced by both the mother and fetus during labor and delivery varies considerably and is likely to be different in primiparous and multiparous women. In this study we explored relations between the birth experience and lactation performance of 40 women. Stress hormones were measured in serum or plasma during pregnancy, parturition (cord and maternal blood), and lactation. Milk samples were obtained and breast-feeding frequency was recorded each day during the first 2 wk postpartum. Four outcomes were used as markers of lactogenesis: the time when the subject first felt fullness in the breasts, 24-h milk volume on day 5 postpartum, milk lactose concentration on day 5, and the day of appearance of casein in the milk as determined by gel electrophoresis. There were significant intercorrelations among the outcome variables, except for milk lactose. Compared with multiparous women, primiparous mothers experienced a delay in breast fullness and lower milk volume on day 5. In multiple regression analyses excluding subjects who had cesarean deliveries (n = 5), markers of both fetal and maternal stress during labor and delivery (cord glucose concentration and maternal exhaustion score) were associated with delayed breast fullness and casein appearance; delayed casein appearance was also associated independently with a longer duration of labor. Milk volume on day 5 was lower in women with higher exhaustion scores, and, in multiparous women, those who breast-fed less frequently on day 2. The milk lactose concentration was inversely related to pregnancy weight gain. These results indicate that primiparity, long labor, stress to the mother and fetus during labor and delivery, and elevated cord glucose concentrations are risk factors for delayed lactogenesis.
Article
(Anesth Analg. 2013;116(2):399–405)
Article
To examine a possible role for gonadal steroid hormones in the enhanced accumulation of hypothalamic oxytocin (OT) messenger RNA (mRNA) and peptide in late pregnancy, we used an established model (22) in which sequential administration of estradiol (E2) and progesterone (P) SILASTIC capsules to ovariectomized rats is followed by removal of P. Long term and sustained E2 combined with abrupt P withdrawal mimics the gonadal steroid hormone pattern of late gestation in the rat (22). Using this paradigm, we demonstrate that OT mRNA is increased in the rat hypothalamus after long term P treatment, but only in the presence of E2 and only when P capsules are removed 48 h before killing. Furthermore, we show that P replacement in primiparous rats during late pregnancy blunts the increase in OT mRNA normally observed at the end of gestation. Our results support a role for E2 priming and P withdrawal in the enhanced accumulation of OT mRNA in the hypothalamus of the female rat.
Article
One of the most fascinating developments in the field of neuroscience in the second half of the 20th century was the discovery of the endogenous capacity of the brain for reorganization during adult life. Morphological and functional mechanisms underlying brain plasticity have been extensively explored and characterized. However, our understanding of the functional significance of these plastic changes is still fragmentary. This book shows that brain plasticity plays an essential role in the regulation of hormonal levels. The second aim is to propose that hormones orchestrate the multiple endogenous plastic events of the brain for the generation of adequate physiological and behavioral responses in adaptation to and in prediction of changing life conditions. The book starts by introducing the conceptual backgrounds on the interactions of hormones and brain plasticity. It then devotes itself to the analysis of the role of brain plasticity in the regulation of the activity of endocrine glands. It examines different hormonal influences on brain plasticity. Then, it goes on to cover the interactions of hormones and brain plasticity along the life cycle under physiological and pathological conditions.