Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

NEUROSCIENCE AND

BIOBEHAVIORAL
REVIEWS

PERGAMON Neuroscience and Biobehavioral Reviews 23 (1999) 673–685


www.elsevier.com/locate/neubiorev

Neurobiology of mother–infant interactions: experience and central


nervous system plasticity across development and generations
A.S. Fleming a,*, D.H. O’Day b, G.W. Kraemer c
a
Department of Psychology, University of Toronto at Mississauga, Mississauga, Ontario, Canada L5L 1C6
b
Department of Zoology, University of Toronto at Mississauga, Mississauga, Ontario, Canada L5L 1C6
c
Department of Kinesiology, Harlow Primate Laboratory, University of Wisconsin, Madison, WI 53715, USA
Received 6 March 1998; received in revised form 19 November 1998; accepted 8 December 1998

Abstract
The optimal coordination between the new mammalian mother and her young involves a sequence of behaviors on the part of each that
ensures that the young will be adequately cared for and show healthy physical, emotional, and social development. This coordination is
accomplished by each member of the relationship having the appropriate sensitivities and responses to cues that characterize the other.
Among many mammalian species, new mothers are attracted to their infants’ odors and some recognize them based on their odors; they also
respond to their infants’ vocalizations, thermal properties, and touch qualities. Together these cues ensure that the mother will nurse and
protect the offspring and provide them with the appropriate physical and stimulus environment in which to develop. The young, in turn, orient
to the mother and show a suckling pattern that reflects a sensitivity to the mothers odor, touch, and temperature characteristics. This article
explores the sensory, endocrine, and neural mechanisms that underlie this early mother–young relationship, from the perspective of, first, the
mother and, then, the young, noting the parallels between them. It emphasizes the importance of learning and plasticity in the formation and
maintenance of the mother–young relationship and mediation of these experience effects by the brain and its neurochemistry. Finally, it
discusses ways in which the infants’ early experiences with their mothers (or the absence of these experiences) may come to influence how
they respond to their own infants when they grow up, providing a psychobiological mechanism for the inter-generational transmission of
parenting styles and responsiveness. q 1999 Elsevier Science Ltd. All rights reserved.
Keywords: Maternal behavior; Brain plasticity; Development; Maternal experience; Olfaction; Somatosensation; Hormones; MPOA; Amygdala; c-fos; Signal
transduction; PKC; CaMBPs; Maternal deprivation; Rat; Monkeys; Humans

1. Introduction towards the mother, seeking and attachment to her teats, and
emission of sound and odor signals that elicit maternal
Non-human mammalian mothers typically display a responses (crying, squealing, urination, and so forth). The
stereotyped set of behavioral responses to their newborn. reciprocal exhibition of maternal behaviors and newborn
They exhibit species-characteristic ways of transporting, signaling promotes physiological and immunological resi-
holding, feeding, and grooming the young, and of protecting lience, physical maturation, and species-typical social and
them from the predators and other dangers [1]. For instance, emotional development in the young (see [2–8]). Ulti-
the prospective mother rat constructs a nest before giving mately, reciprocal mother–infant behaviors increase the
birth. She aids the birth by pulling individual pups from the probability that the young will survive and, beyond survival,
vaginal canal, severs the umbilicus, eats the placenta, cleans will mate and successfully rear their own offspring. In this
the pups, and carries them one-by-one in her mouth to the article, we show how these behavior patterns, previously
nest. In the nest, she continues to lick the pups, especially viewed as fixed and stereotyped, are in fact quite plastic,
their anogenital regions, and eventually adopts a nursing and depend on brain structure and function that was substan-
posture over them. In return, mammalian neonates exhibit tially influenced by experience at the time of birth and over
behavior that is even more stereotyped and rigid than mater- the life-span.
nal behavior. This includes orchestrated orienting responses As shown in Fig. 1, the development of the capacity to
both express and modify maternal behavior patterns in
* Corresponding author. Tel.: 11-905-828-3961; fax: 11-905-569-
adulthood may depend on mechanisms that were themselves
4326. activated and later tuned by early experiences. The interac-
E-mail address: afleming@credit.erin.utoronto.ca (A.S. Fleming) tion between the newborn and the mother alters basic
0149-7634/99/$ - see front matter q 1999 Elsevier Science Ltd. All rights reserved.
PII: S0149-763 4(99)00011-1
674 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

Fig. 1. Schematic showing factors that affect the newborn infant’s response to the mother and mother’s response to infants. In this ‘Intergenerational’ model,
we describe the hypothesized transmission of experiences received by the infant (F1 generation) in the nest and in relation to their own mothers on the quality of
mothering the offspring show towards their offspring (F2 generation) when they grow up. The assumption is that the transmission of experience effects persists
across generations, from generations F1 to F2 to F3, etc. Influences experienced by the developing offspring are determined in part by their genetic makeup, by
uterine experiences, by hormones (both pre- and post-natally), and by present context in the nest and with mother and/or siblings. Experiences alter the
psychobiology of the offspring, changing their subsequent neuroanatomy, chemistry, endocrinology, and, hence, behavior. These offspring, then grow up and
gain experiences throughout their lives which, together with their earlier experiences, will determine how they respond to their offspring.

mechanisms of behavioral expression in both. How the sheep mothers, ewes), mothers rapidly learn to identify
mother responds determines, in part, how the neurobiologi- their own infants based on their odor characteristics. In
cal and behavioral changes in the infant proceeds. Changes species that do not show selectivity (e.g. rats), the early
in an infant may eventually play out as to how the individual postpartum experiences with the litter helps to maintain
will respond to the offspring as an adult, and so on. maternal responsiveness to young during subsequent lacta-
tion. Although processes of learning are likely to be
involved in both types of experience, the mechanisms
2. Reciprocity in the mother–young relationship mediating mothers’ recognition of individual infants are
somewhat different from those that mediate the effects of
2.1. Mothers learn about and from their infants: role of experience on maternal responsiveness in general.
sensory, hormonal, and neural influences
2.1.1. Maternal experience affects individual recognition of
Experiences acquired while mothers are with their infants offspring
influence their willingness to care for the young. In many Under the influence of parturitional hormones ewes who
primate species (including humans), prior experiences of have just given birth may initially nurse any lamb, but they
holding and carrying the young either during adolescence soon learn to recognize their own lamb by its odor [15].
(siblings) or after a previous birth can have profound effects Once an ewe has smelled her lamb’s odor (usually the
on how competent or motivated females care for their mother’s own amniotic fluid), she butts away alien-smelling
offspring (humans, [9]; marmosets, [10], rhesus macaques, lambs and permits to nurse only her own lamb. Her recogni-
[11–13]; vervet monkeys, [14]). In some species, absence of tion requires only a few minutes of exposure to the lamb to
pre-pubertal experience with younger siblings results in form [15]. In humans also, new mothers come to recognize
first-time mothers who neglect or even eat their infants their own infants’ odors, cries and tactile characteristics
[10]. Postpartum maternal experience can also affect [16–18]. They do so even with very brief exposure to the
mothers’ subsequent responsiveness to these same offspring features of the infant during the first few postpartum days.
at a time when the hormones of parturition are no longer Mothers who nurse their infants sooner after birth and/or
behaviorally effective. In species that respond selectively to who keep their faces in close proximity to their infants while
their own infants, to the exclusion of other infants (e.g. holding them (hence acquiring more olfactory experience)
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 675

are both more attracted to their infants’ body odors and hormonally regulated. Once the maternal behavior has
come to recognize them more easily [9]. been exhibited it persists for a number of weeks in the
We now have a substantial understanding of the endo- absence of parturitional hormones [28,29]. Hence, for the
crine mechanisms that mediate the enhanced responsiveness new mother, care of offspring appears to depend on hormo-
to offspring normally seen in the mother at parturition (see nal activation of neural mechanisms that express maternal
[1,19,20]). Endocrine and neurochemical mechanisms regu- behavior. Once this has happened the hormones are no
lating the expression of maternal behavior intersect or over- longer required.
lap with those that activate or prime discriminate learning Pup stimuli have intrinsic effects on the female nervous
mechanisms. In most mammals that have been studied, the system. Virgin rats can be induced to become maternal
hormones that activate maternal responsiveness to young in through repeated exposure to pups over five to seven days
general are those that change at the time of parturition. [30]. Maternal behavior is exhibited most rapidly towards
These include a period of elevated progesterone during pups when the parturitional hormones are present, however.
pregnancy, followed by its pre-parturitional decline When interacting with the young, the hormonally influenced
[20,21,106]; a late pregnancy rise in circulating estradiol maternal rat associates how the pups smell, feel, and sound
and pituitary prolactin; and a rise with parturition in oxyto- with motor responses involved in picking up and moving
cin [20,21]. In ewes, a variety of hormones and neuropep- pups and adopting a crouch posture over them. Through
tides including estradiol, oxytocin, endogenous opioid concurrent hormone changes and exposure to pups, the
peptides, and corticotrophin releasing hormone (CRH) are mother becomes habituated to their odors and other novel
elevated at parturition and act in synergy with cervical aspects that would usually activate fear and withdrawal
stimulation (which occurs during the birth of the lamb and [31,32]. Unlike mother sheep who learn the individual
stimulates oxytocin release) to enhance the learning of the odors of their lambs, rodent mothers do not develop indivi-
new lamb’s odors [15]. The catecholamines norepinephrine dual pup recognition. They respond to their own and other
(NE) and dopamine (DA), along with glutamate and g- litters of the same age in much the same way. Parturitional
amino-butyric acid (GABA) and nitric oxide are also hormones enhance the attractiveness of pups and their odors
released or synthesized within the olfactory bulbs at this [31] and facilitate maternal learning [33]. Pups become
time. In addition, levels of brain NE at parturition are higher considerably more reinforcing to the dam once she has inter-
in multiparous than in primiparous ewes [22]. Hence, the first acted with them and become experienced. In fact, mothers
maternal experience may prime (or sensitize) the neurochem- exhibit preferences for visual environments that have been
ical mechanisms regulating the formation of specific discrimi- associated with pups in only two trials [34]. Maternally
nation of subsequent offspring. At parturition, release of these experienced females show enhanced bar-pressing for deliv-
neurochemicals facilitates changes in brain mechanisms ery of pups by comparison to the inexperienced females
underlying the recognition process [15,22–24]. [35].
Consolidation of the association between the mother’s As with other types of learning, the integrity and resili-
birthing stimuli (cervical dilation) and attraction to the ence of maternal learning/memory varies with frequency
lamb’s odor appears to involve changes in the olfactory and duration of exposure and the interval between exposure
bulb cytoarchitecture and synaptic efficiency [22–24] and and test. If new mothers are separated from their young and
possibly, amygdala, and hypothalamic processes as well. not permitted to interact with them at all, their maternal
The importance of these limbic and hypothalamic sites for responsiveness declines over the first postpartum week.
the onset and maintenance of maternal behavior is most By 10 days postpartum, the mother withdraws from the
evident in the research on rodents (see the following). foster pups much as the naive females would [36]. However,
if permitted as little as two hours of contact with the pups on
2.1.2. Maternal experience affects maternal responsiveness day 1 postpartum, the mother rat shows very active maternal
In contrast to the many species that require early pre- response to the foster pups presented 10 days later [36].
pubertal or subadult caretaking experience to show adequate Initial maternal learning depends primarily on somato-
mothering, female rats under laboratory conditions, and sensory and chemosensory modalities. If new mothers are
with no prior experience with rat pups, usually care rendered anosmic and cannot smell pups and are also unable
adequately for their first litter. Female rats who have not to acquire somatosensory stimulation of the ventrum and
given birth usually withdraw from or avoid pups, so changes mouth regions (due to application of anesthetic onto or
surrounding the birthing process must alter this pre-potent into these regions), pups are not licked or retrieved
response. The inexperienced mother rat builds a nest, gives [37,38]. Maternal olfactory and somatosensory processing
birth, and then gathers, licks, and nurses her young under the is necessary for pups to acquire reinforcing value, and for
influence of changing levels of circulating steroidal and the mother to retain responsiveness to pups over long peri-
protein hormones (see [1,20]). The hormones associated ods of separation [37,39]. As an example, presenting
with lactation may also contribute to the patterning of mothers with pups scented with an artificial odor results in
nursing after parturition [27]. However, the later persistence preferential attraction to that odor in an odor preference task
of a motivation to care for the young seems not to be and preferential maternal treatment of similarly scented (as
676 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

Fig. 2. Functional neuroanatomy mediating maternal and related behaviors in mammals. Neuroanatomical structures include olfactory bulbs, amygdala
(medial (Me), cortical (CORT), central (Ce), and basolateral (BASOLAT), nuclei, nucleus accumbens, bed nucleus of the stria terminalis (BNST), medial
preoptic area, midbrain, and parietal cortex. Relevant neurochemistry includes the catecholamines, norepinephrine (NE), and dopamine (DA), the neuropep-
tides, g-amminobutyric acid (GABA), N-methyl-d-aspartate (NMDA) and opioids.

opposed to ‘otherly’-scented) pups in tests 10 days later. In mechanisms and we know some of the neuroanatomical
order to continue the caring of pups after the hormonal systems underlying aspects of general learning per se, a
effects have subsided, the mother must continue to find next step was to determine the role of these brain structures
them attractive. Hence, mechanisms of reinforcement, and those in the ‘maternal circuit’, in the acquisition and
learning and memory are recruited. Hormones may facili- retention of maternal experience. For instance, work on the
tate the relationship, but learning factors maintain it. Facili- supraoptic nucleus shows that, with suckling experience in
tated postpartum learning also occurs in olfactory-social the lactating animal, there occur changes in the structural
contexts which are not related to pups, indicating that the and functional properties of supraoptic neurons. These
general learning processes are recruited [40]. neurons release oxytocin in response to suckling stimulation
The combined role of hormonal changes and sensory and are necessary for the milk letdown reflex. [44]. Modney
experience in promoting maternal behavior and learning is and Hatton [44] found two kinds of change associated with
fairly well understood [9,29]. Until recently, however, there the end of pregnancy and the initiation of nursing. One
has been virtually no understanding of which neuroanato- involves the formation of new (double) synapses on the
mical and neurochemical systems are responsible for the cell body and dendrites of the supraoptic nucleus, while
experiential modification of maternal behavior. One the other involves the increase in electrical coupling
approach was to investigate the neurochemical systems among these same oxytocinergic supraoptic neurons.
that are known to be involved in the mediation of learning These changes very likely contribute to the synchronized
and memory in other functional contexts that involve affec- activity in these oxytocin cells prior to milk ejection. Of
tive changes. As with other types of learning, maternal particular interest in this context are the additional findings
learning requires protein synthesis for its consolidation. that some of these modifications also appear in virgin
Injection of protein synthesis inhibitors (e.g. cyclohexi- animals induced to become maternal through long-term
mide) into the ventricles of the brain after exposure to exposure to pups, but who did not receive suckling stimula-
pups for three hours blocks the long-term retention of the tion. Instead, there is evidence that olfactory and/or other
behavior [41]. NE mechanisms are also involved as b-nora- chemosensory stimulation derived through licking and
drenergic blockers administered after this exposure block retrieving of pups may induce these changes in the virgin
the long-term effects whereas isoproterenol, an NE agonist, [44,45]. The time course of these effects in the nursing
enhances the retention [42]. Finally, the development of animal may be relevant; some synaptic changes are rever-
maternal conditioned place preference using pups as rein- sible, but apparently others are more permanent. Such long-
forcing stimuli is blocked by a-flupenthixal, a DA antago- term structural changes could provide a possible basis for
nist [34]. Our particular interest, though, is in the relation the findings that, once dams have had some nursing experi-
between learning systems and maternal systems [29,43]. ence, distal olfactory and other exteroceptive pup cues come
As there is a considerable overlap of maternal learning to elicit the conditioned release of oxytocin in the absence of
with more general conditioning and learning reinforcement suckling [46].
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 677

bed nucleus of the stria terminalis, the medial, cortical


and basolateral nuclei of the amygdala, the nucleus accum-
bens, the olfactory and somatosensory cortices, and the pre-
frontal cortex [51,52]. Fig. 2 shows the functional neuroa-
natomy mediating the maternal and related behaviors. Some
of these sites are clearly part of the final common path for
the expression of maternal behavior (e.g. MPOA,
[29,43,55]). Others mediate stimulus salience and effect
(medial and cortical amygdala, see [56,57]), and processes
of learning, memory and reinforcement (pre-frontal cortex,
basolateral amygdala, nucleus accumbens, see [51,58]).
Others participate in sensory processing (cortical and
medial amygdala, parietal and olfactory cortices [56,59].
Only a few of these sites are activated at retention testing.
When re-exposed either to pups themselves or to pup-asso-
Fig. 3. Potential signal transduction pathways leading to maternal c-fos ciated conditioned cues (an environment or stimulus paired
expression. Maternal hormones, neurotransmitters and pup stimulation (e.g. with pups), the only sites that are more active in the experi-
olfactory) can activate many receptor complexes. This leads to the eleva- enced than the inexperienced animal are the MPOA, the
tion of neuronal calcium levels and the stimulation of protein kinase C
(PKC). Calcium activates calmodulin (CaM), which in turn stimulates a
basolateral amygdala and the parietal cortex [50]. Experi-
wide range of calmodulin binding proteins (CaMBPs) such as CaM-depen- ential effects of performing maternal behavior and receiving
dent kinases. These kinases and PKC each phosphorylate specific upstream pup stimulation in the postpartum mother are the basis for
elements that ultimately lead to the expression of c-fos. As part of transcrip- maintaining maternal behavior when hormonal effects have
tional complexes, such as AP-1, c-fos could then activate gene expression waned. MPOA lesions produce severe deficits in both the
that underlies maternal behaviour.
hormonal and experiential components of maternal behavior
[43]. The transition from hormonal to non-hormonal regula-
Using quite a different approach we used the c-fos immu- tion of maternal behavior, therefore, is probably dependent
nocytochemistry to assess which brain areas are activated on MPOA mechanisms.
during the acquisition and retention of a maternal experi-
ence.
4. Effects of experience on signal transduction
components
3. Correlates of maternal behavior at the intra-cellular
level Although c-fos expression is clearly enhanced with the
expression of maternal behavior and experience, it is not
3.1. Fos as a neuroanatomical marker of maternal known whether this protein is necessary for the behaviors
experience to be expressed and, if so, what role they play. As our
interest lies in understanding the regulatory events that initi-
Fos is a protein produced by a class of immediate early ate maternal behavior and experience, we have started to
genes known as c-fos oncogenes that are present in brain determine how c-fos expression is regulated by up stream
neurons and activated by a variety of stimulus situations and regulators of maternal behavior, and to determine what
behavioral states [47]. Acting with another proto-oncogene function the expressed proteins serve. In mammalian cells,
protein, Jun, as part of the AP-1 transcription factor, Fos can there are numerous signaling pathways that can lead to c-fos
operate directly as part of the transcriptional apparatus to expression [60]. One approach to dissect the major signaling
direct gene transcription or it can act by binding to enhancer pathways in specific regions of the rat brain is to immuno-
elements to affect the gene regulation [48]. Therefore, c-fos localize selected proteins from those pathways, and then to
immunocytochemistry can be used as a tool to determine determine if they are present during and are linked to the
which brain sites are activated when an animal first responds maternal behavior. Two major components of calcium-
maternally, acquires an experience, and then retrieves the mediated signaling pathways that lie upstream to c-fos
memory [43,49–52]. Moreover, the physiological relevance expression were examined: protein kinase C (PKC) and
of Fos for maternal behavior is suggested by the recent calmodulin binding proteins (CaMBPs; Fig. 3). At least
observation that c-fos-knockout mice fail to show adequate two of the protein hormones involved in the expression of
maternal behavior under circumstances in which it would maternal behavior, oxytocin and prolactin, can produce
normally be seen in the wild-type mouse [53]. intracellular neuronal increases in calcium levels, leading
If an animal is maternally active for the first time, to the activation of PKC and CaMBPs [61]. PKC can be
enhanced c-fos expression is found in a number of brain activated by diverse signaling pathways, including via G
regions including the medial pre-optic area (MPOA), the protein-linked phospholipases, receptor tyrosine kinases
678 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

and non-receptor tyrosine kinases [62,63]. While numerous has further revealed the absence of calcineurin from the
PKC forms of the protein exist (isozymes), the brain-speci- MPOA but not other regions of the maternal brain (O’Day
fic PKC isozyme (PKC gamma) is a typical C kinase that is et al., unpublished results). These changes and differences
activated by calcium ions and diacylglycerol and undergoes are localized only to the MPOA suggesting that there may
developmental increases in the rat brain [63]. PKC is impli- be calcium-regulated signaling pathways that are specifi-
cated in long-term potentiation (a neuronal model of cally associated with maternal behavior and plasticity. The
memory) as well as in learning, memory, and plasticity goal now is to characterize these CaMBPs to gain further
processes [64,65], PKC can also act as an upstream regula- insight into their possible functions.
tor of AP1 (c-fos/c-Jun) expression [48,66].
Immunocytochemical assay of a number of different PKC
isozymes indicates that there is a considerable overlap in the 5. A functional neuroanatomy of maternal experience
regional and temporal pattern of expression of PKC and Fos
protein in mother rats exposed to offspring [67]. There are These studies not only reveal some of the basic molecular
also some incongruities in their patterns of expression that mechanisms underlying maternal processes, but also help to
may be informative. For example, enhanced staining of both elucidate the neuroanatomy that underlies the acquisition
PKC and Fos protein is observed in the MPOA and the and retention of maternal experience (see Fig. 2). A role
parietal cortex after maternal experience with pups [32]. If for the MPOA, the limbic system, the olfactory and soma-
the pups are removed, enhanced PKC will still be observed tosensory systems in the initiation and retention of maternal
in the mother four days later but Fos staining would have behavior is consistent with more general learning, memory,
diminished by then. Active stimulation by pups or pup-asso- and emotional functions of these structures.
ciated conditioned cues is necessary to reinduce enhanced c- Beyond this it is known that the MPOA contains receptors
fos expression, and this occurs rapidly but is also relatively for the hormones that activate both the motoric and motiva-
short-lived. The relation between the PKC and Fos results tional aspects of maternal behavior [28,55]. Lesions of the
suggest that maternal experience initially stimulates MPOA eliminate maternal retrieving and nest-building as
enhanced PKC synthesis and an activation of the c-fos well as instrumental responding for pup-delivery in an oper-
gene in the maternal system (e.g. MPOA). In the absence ant apparatus [35,43]. Electrical stimulation of this area has
of pup stimuli, maternal PKC remains elevated for a consid- opposite effects [55]. Of all the brain structures studied from
erable period, but c-fos gene activation declines. As PKC a molecular perspective, the MPOA undergoes the most
sits upstream in the biochemical cascade leading to c-fos robust and substantial change with maternal experience.
activation, enhanced PKC resulting from past experience MPOA function is substantially altered by experience, and
with pups would permit rapid activation of the c-fos gene such alterations may play a role in the facilitated maternal
in relation to new pup stimuli in the experienced female. behavior seen in the experienced animals. The hormonal
Another primary route for calcium function is via calmo- responsiveness of the MPOA increases with increasing
dulin (CaM) and theCaMBPs to which it binds and modu- parity [21]. Moreover, electrical stimulation of this brain
lates. Upon activation by Ca 21, CaM was shown to regulate site is more effective in promoting maternal behavior in
c-fos expression but the specific CaMBPs that mediate this experienced as opposed to inexperienced animals [55]. In
expression have not been identified [68]. However, the short, it appears that the MPOA is involved not only in the
CaMBP, CaM-kinase II has been implicated in models of initial expression of maternal behavior, but also in retaining
neuronal plasticity [69]. In addition, certain kinases and and mediating the effects of maternal experience and parity
calcineurin (CaM-dependent protein phosphatase 2B), a on the subsequent expression of the behavior.
major brain protein, regulate transcription factor phosphor- Other central nervous system structures clearly mediate
ylation upstream of Fos expression [70]. Cells also contain the sensory aspects of maternal behavior (e.g. parietal cortex
dozens of CaMBPs many of which mediate neurotransmitter and ofactory bulbs), being activated when the animals sniffs,
release [71]. Previous studies have identified only a handful licks, mouths and crouches over the pups [38]. The parietal
of the many CaMBPs in brain and other tissues of the rat. cortex also appears to be changed by maternal experience,
Rather than focusing upon one or two known CaMBPs, an effect that is consistent with evidence that this sensory
analysis of the pattern of changes in the total population receiving area is plastic and re-organizes in relation to
of these proteins during maternal behavior has been a fruit- experience in adult rodents. For example, in comparison
ful approach in elucidating the role of these important cellu- to the non-lactating postpartum females, lactating (and
lar regulators. A gel overlay technique with recombinant hence more experienced) females have an expanded region
calmodulin (35S-VU1-CaM) reveals over two dozen of the cortex activated by stimulation of the ventral teat
CaMBPs, a number in keeping with the known population region [73]. What this probably means is that cortical
extant in mammalian and other eukaryotic cells [72]. Analy- mechanisms change in relation to experience; and subse-
sis of the quantitative changes in the CaMBP profiles quent experiences are almost certainly processed differently
reveals that there is a decrease in several CaMBPs asso- than were the initial experiences.
ciated with the onset of maternal behavior. Western blotting A role for amygdaloid nuclei in the expression and
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 679

retention of maternal behavior has alsobeen demonstrated. 6.1.1. Short-term experience effects
Areas in the medial and cortical amygdala receive olfactory For instance, rat pups develop an attraction to the chemo-
input and are believed to mediate avoidance responses to sensory characteristics of mother’s ventrum and first
odors or have anxiogenic actions more generally. Indeed, attaches to the nipples based on prior intrauterine experi-
these amygdalar mechanisms inhibit the expression of ence with the amniotic fluid [76]. Attraction is often
maternal behavior in virgin animals. Lesions of these nuclei followed by the development of nipple-preferences. In
or of pathways that project from the amygdala to the MPOA fact, kittens develop a preference for a particular nipple or
in the virgin animal permit rapid maternal responding to nipple location on the mother’s ventrum. Seeking the ideal
pups and reduce fear responses to odors in other contexts nipple position is based on experiences with the tactile and
(see [9,32,43,55]). thermal characteristics of the mother’s ventral surface [77].
The enhanced activation of the basolateral amygdala with In rats, the expression of food preferences is based on
maternal experience is consistent with the observation that experiences acquired during the infant’s exposure first to
this structure receives extensive primary or secondary input the mother’s milk, then her odors (which are partially
from all the relevant sensory modalities, thereby serving as a dependent on her diet), then to food particles remaining in
point of convergence where the formation of associations the vicinity of the mother’s mouth and on her fur, and
may occur [52,56,59,74]. Walsh et al. [52] demonstrated finally to following the mother to a food site [78–81].
that the combined chemosensory and somatosensory desen- More subtle discrimination by weaned animals of nutri-
sitization in maternally-behaving animals produces an addi- tious versus nutritionally deficient foods is also based
tive reduction in basolateral amygdaloid activity (reflected on learning [82].
in c-fos expression). Moreover, lesions of this region elim- The behavioral effects of early experiences occurring in
inate the development of a conditioned place preference for the maternal nest are mediated by neurochemical and
pups and disrupt the postpartum expression of maternal neuroanatomical mechanisms that have been substantially
behavior [35]. Not surprisingly, given its role in reinforce- changed by early experiences [2–3,83–85]. As an example,
ment [58,75], DA depletions or lesions of the nucleus the recognition of the rat mother’s specific odor by pups
accumbens also disrupt the consolidation of maternal occurs in conjunction with licking of the pups by the mother
memories [28]. Hence, the combined activation of basolat- [86]. Formation of the association between licking and
eral and nucleus accumbens in response to pup stimuli, and maternal odor depends on licking induced activation of
the effects of lesions in reducing the maternal responsive- the midbrain nucleus, the locus coeruleus (LC), whose
ness, suggest that the basolateral amygdala and nucleus axons terminate in the olfactory bulbs and release the neuro-
accumbens form an interconnecting neural loop that transmitter, NE [86]. Recognition of maternal odor involves
mediates the formation of associations between biologically both the neurochemical and cellular cytoarchitectural
relevant events (pup stimuli and maternal behavior, in this changes in the olfactory bulb [86,87]. Development of an
instance) [56]. attraction and preference for maternal odors (over other
The effects of maternal experience on the basolateral odors in the environment), depends on central processing
amygdala and on the parietal cortex reflects the general of olfactory information by the limbic structure, the amyg-
properties of these two structures (systems) that are dala. More generally, processing of the olfactory bulb inputs
recruited by many types of learning. Experience-related by amygdalar mechanisms determines the salience of olfac-
activation of MPOA seems to be specific to maternal beha- tory stimuli and the nature of affective responses to them
vior, however. Perhaps it makes sense that in mammals, as [86]. The neuropeptides (oxytocin and opioids) may also
in birds [4,6], the long-term effects of memory should be play a role in the infant rat’s early affiliative behaviors in
most clearly seen in the very system that normally underlies the nest and in the infant’s attraction to maternal odors [25–
the expression of the behavior. 26,85,88]. The proclivity of rat pups to approach and attach
to the mother is heavily dependent on the experiential
effects on olfactory and limbic mechanisms that mediate
attraction to and recognition of the mother and her environ-
6. Intergenerational effects: Does how one was mothered ment.
influence subsequent mothering?
6.1.2. Long-term experience effects
6.1. The infant becomes experienced through the mother Postpartum experiences with the mother and nest influ-
ence the development of the infant prior to weaning.
Among mammals, the maternal and nest contexts provide Concurrent early olfactory experiences (mothers’ odors)
a rich learning environment for the developing young. and somatosensory experiences (being nursed, licked, etc.)
Mothers not only learn about their offspring; but the also appear to have long-term effects on the infants as they
offspring also gain experiences during the pre-weaning grow. For instance, infant rats who received more touch and
period that affects their adjustment to the mother and later licking stimulation from the mother in the nest show a
on to their own offspring. higher level of pup-licking as juveniles and as adults
680 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

when presented with new-born pups, by comparison to less together based on findings of multiple short-term studies
stimulated infants; moreover, recent studies indicate that the and there are missing pieces to the puzzle.
quality of the infants’ nest experience may well affect their What has been investigated is the relationship between
later behavior with their own offspring (Natterer, Lovic, maternal and infant attachment style, and later personality
Shah, and Fleming, in preparation). Similar early maternal of the infant-as-young-adult and attachment style of their
stimulation in the nest also produces a dampening of the infants. The results of short term longitudinal studies indi-
offspring’s emotional reactivity to novelty and stress when cate that (1) the emotional quality of mothers’ responsive-
they become adults [89]. Reductions in stress reactivity are ness to infants is related to the infants’ attachment style, (2)
attributable in part to the increased density of hippocampal infants’ attachment style in turn is related to social and
glucocorticoid receptors. These receptors normally mediate affiliative aspects of adult personality, and (3) these adult
negative feedback effects of circulating adrenal glucocorti- personality styles are related to responsiveness to infants
coids following hypothalamic-pituitary-adrenal (HPA) acti- [92–94].
vation. Stimulation-induced changes in thyroid function and Scientific interest in intergenerational effects has also
brain serotonin system activity early in the postpartum been driven by efforts to understand the mechanisms under-
period are also involved in these long-term hippocampal lying the abnormal by comparison to normal psychosocial
effects [90]. Hence, long-term changes in brain mechanisms development. In social primates, the importance of early
that modulate the stress reactivity in offspring are produced mothering in determining how the daughters will mother
by natural variations in mothering behavior. To the degree is clearly demonstrated under the conditions in which
that many interactions with offspring are stressful, this may female offspring are partially or completely deprived of
canalize some aspects of the way individuals respond to early mothering. For instance, rhesus monkeys can be reared
their own offspring later on. In species in which mothers with same aged peers rather than mothers (peer-reared
develop individual recognition of their offspring, as in monkeys) [79,95–98]. When peer-reared monkeys become
sheep, the same or similar mechanisms used to promote mothers as adults, they are more likely to reject or abuse
attachment of the young female to her mother, are re-acti- their infants than monkeys reared by their biological
vated when she gives birth and exhibits recognition and mothers (mother-reared monkeys). The mechanisms that
attachment to her own offspring. This transfer of biological mediate this effect are not understood. The idea that peer-
effects of early rearing experience to later maternal respon- reared monkeys are generally poor mothers because they are
siveness and behavior may be especially evident when deprived of ‘learning how to mother’ from their own mother
considering species in which the parent is intimately cannot entirely explain their deficits in maternal behavior.
involved with disciplining or socialization of offspring, as Some peer-reared monkeys are perfectly adequate mothers,
in non-human primates. but the proportion of who are not considerably exceeds that
In rhesus monkeys, measures of HPA axis activity in observed in mother-reared monkeys. As no peer-reared
mothers and offspring are positively correlated [91]. In monkey has experience with maternal care, the range of
vervet monkeys, there is substantial evidence that the style variation in peer-reared maternal behavior cannot be
of mothering exhibited by adult daughters is similar to the explained by early variation in learning how to mother.
style of mothering shown by their mothers [14]. Vervet In general, how the individual copes with environmental
monkey mothers who engage in a high level of mother– change and stress may vary with how they cope with chal-
infant ventral contact have daughters who also show high lenges presented by offspring. By comparison to mother-
mother–infant contact. Abusive patterns of maternal care reared monkeys, peer-reared monkeys are behaviorally
span generations as well, and the incidence of abuse varies over-reactive to stressors such as competitive social inter-
across matrilines in pigtail macaques [14]. One cannot, of actions or forced separation from their social group [79,95–
course, identify the mechanism underlying the intergenera- 98]. The peer-reared monkey is both more likely to be inor-
tional similarity based on these correlations. Similarity dinately aggressive in social altercations, and more inactive
could be due to consistent (genetically pre-programmed) and withdrawn when isolated apart from its accustomed
personality styles or due to the effects of observing the social group. They exhibit chronically lower levels of
mother’s interactions with younger siblings. Using multi- brain catecholamine (CA) system activity, but then have
variate analyses in which these alternate influences were exaggerated increases in brain CA activity when stressed
assessed, however, Fairbanks [14] demonstrated that the [79,95–98]. Their HPA axis activity is also chronically
manner in which the infants were mothered did indeed reduced, somewhat paradoxically, in relation to what
exert an effect on their adult mothering behavior. Among seems to be CA system hypersensitivity, their HPA axis
humans, it is commonly assumed that there are often inter- response to stressors is blunted [95]. In addition, there are
generational similarities in maternal behavior. We have not lawful relations between neurochemical and neuro-
yet found a longitudinal study that conclusively documents endocrine levels and behavior in mother-reared monkeys,
substantial and significant commonalties in specific prac- and these relations are less prominent or simply absent in
tices of mother–daughter maternal behavior, however. juvenile peer-reared monkeys [97]. Hence, in rhesus
Rather, the long-term longitudinal story has to be pieced monkeys, one persisting effect of maternal deprivation is a
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 681

neurobiological disorganization which is accompanied by a recognition, attachment, and caregiving? Plasticity permits
relative inability to cope behaviorally or physiologically a certain latitude or flexibility in the relevant features that
common social stressors later in life [99]. Among such stres- enable the offspring to identify the mother, and for the
sors would be caring for offspring, and as noted before, mother to identify her offspring. On the one hand, olfactory
peer-reared mothers are substantially more likely to reject cues and other characteristics of the rodent mother may vary
or abuse their infants. depending on her sources of food and dietary regimen.
Although the neurobiological effects of complete mater- These resources vary geographically, and over days,
nal deprivation have not been studied in humans, it is known weeks, and seasons, and consequently are not necessarily
that children reared in orphanages in Romania exhibit the same for all of her litters or even pups within a litter.
disrupted physiological, sensory-motor, emotional, and Hence, the pup’s expression of attachment behaviors cannot
cognitive development reminiscent of that observed in be locked on a pre-determined and invariant set of salient
socially isolated rhesus monkeys [98,103]. There is also maternal characteristics. On the other hand, variation in
substantial evidence that infants who received abusive or maternal diet and environmental effects on maternal pre-
neglectful parenting may face difficulty in parenting their parturitional physiology, may affect infant odors, body
own offspring. Approximately 30% of children who were size and conformation, and neonatal behavior. Successive
abused or neglected during their early lives come to abuse offspring will differ in genotype as well as phenotype. If
and/or neglect their own children many years later (versus a expression of maternal behaviors were narrowly locked to
base rate of 5% for non-abused children, see [104,105]). The invariant neonatal characteristics, then the diversity of
intergenerational effect may be that children who have been offspring nurtured and thus surviving would also be narrow.
abused develop personalities as adults, lacking in self- Hence, plasticity in mammalian mother–infant behavior: (a)
knowledge and empathy for their own children’s pain and allows for offspring to be nurtured in widely varying envir-
misfortune, and also fail to cope with the many challenges onments and circumstances, and (b) provides for preserva-
presented by the child. These effects are probably reduced or tion of genetic/behavioral diversity in those offspring. Also,
prevented if the abused child subsequently, or concurrently, by using conditioning mechanisms, a complex multimodal
has affiliative social experiences with a caring individual. stimulus (say, of an infant, who has a characteristic odor,
This is likely to be the reason why 70% of abused children cry, and visual appearance) comes to be easily located and
do not go on to be abusive themselves as parents [100,104, identified based on any one of these cues. This is important
105]. when not all the cues are present (as when the infant is
Based on our understanding of experience effects on brain concealed within a nest or strays away from the nest or
in other animals, it is also possible that children who have mother).
been severely neglected or abused have experienced neuro- Finally, plasticity in the maternal brain increases the
logic changes which result in altered affective, perceptual reproductive fitness by allowing experience with one infant
and cognitive function during development. Such changes to modify the care of subsequent offspring in the changing
are more than likely to affect how they perceive and respond environment in which the mother lives. Among many
maternally to their own offspring, how they approach mater- primate species, mothers of first infants are considerably
nal care responsibilities initially, and how they are able to less competent and motivated than are mothers of subse-
learn or modify maternal care practices. Such conclusions quent offspring. The experiences of the multiparous female
may not be controversial, or not as controversial as they are not limited to interactions with individual offspring.
would have been decades ago when maternal behavior They include somatic and physiological experiences asso-
was generally accepted as a hard-wired aspect of female ciated with pregnancy and parturition, as well as exposure to
brain function. The major theoretical issue then is under- infants, peers, and the larger environment and society, more
standing why this aspect of brain function which is critical generically. This effect of parity and the associated experi-
for species survival is so malleable. ences usually insures that the young of an experienced
mother have an advantage in comparison to the first-born,
the young of less experienced mothers. They are more likely
7. A functional analysis: plasticity and development of to be cared for during adversity and receive more abundant
species-typical behavior nutrition, attention, and social stimulation provided by an
older, more mature, less timid, and better socialized mother.
Plasticity in behavioral mechanisms is not unique to The mother and young usually constitute for one another the
mammalian mother–young interactions, but is exhibited earliest and most pervasive stimulus set present in the post-
across species and in a number of developmental contexts partum environment. To the degree that the nervous system
(e.g. song-learning, food caching, food preference develop- of the infant is dependent for its development on an
ment, imprinting, mate selection [4,5,7,8,82,101,102]. What extended series of interactions with a mother, deprivation
might be the adaptive, and hence selective value of plasti- of these interactions might have adverse effects in the
city and dependence on learning, rather than ‘hard-wiring’ proportion. Plastic brain mechanisms take time to adapt,
of innate behavioral control mechanisms for mother–infant and their development can be adversely affected if they
682 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

are not exposed to the expected or common set of species- subsequent offspring. Other effects of some of the same
specific experiences. early experiences (e.g. receiving adequate somatosensory
stimulation) are more general, leading to different patterns
of response to stressors and social stimuli. These behavioral
8. Summary and future directions effects of experience are mediated by changes in brain
neurochemistry, structure, and function in both the mother
In this article, we have described the importance of learn- and the young.
ing and plasticity mechanisms in the regulation of the It is important to learn which biochemical pathways are
mother–young relationship. In mammals, the attraction operational in the appropriate brain regions and are tempo-
between mother and young, and their mutual recognition rally linked to the onset and consolidation of maternal beha-
depends on olfactory and somatosensory experiences. vior. Any complex behavioral process, such as maternal
Some effects of early experience are remarkably specific, behavior is unlikely to be mediated by a single molecular
leading to recognition of, and attachment to, the mother and entity such as a specific PKC isozyme. Only by knowing the
siblings, and development of spatial location and food functional pathways that are operational during the process
preferences. There is a striking similarity between mechan- will it be possible to understand the potential interconnec-
isms mediating the rat pup’s recognition of maternal odors tions and cross-talk between pathways that current research
on one hand, and the ewe’s recognition of her lamb’s odors is revealing as standard operating procedure for mammalian
on the other. Recognition of the mother by infant and the cells. A complete understanding of these functional path-
infant by mother involves the formation of an olfactory ways is also essential for interpreting the results of genetic
recognition in association with somatic stimulation (cervi- alterations of specific components of the signaling routes.
cal stimulation-ewe, or being licked, nuzzled-pup). Both At a different analytic level, learning and plasticity within
these involve somatosensory activation of the centrifugal the mother–young context play a critical role in promoting
Locus Coeruleus NE system. This activation promotes the adaptive fitness in mammalian species per se, and this role
formation of cytoarchitectural and synaptic changes leading may be extended to non-human and human primates. In
to both an increased discrimination of odors, and an asso- primates, early experience and associated brain changes
ciation of odor information with somatosensory stimulation clearly affect how the infants interact with the world in
provided either by birthing (in the case of the mother), or by general, and with their own offspring as adults. Establishing
maternal grooming or cleaning (in the case of the offspring). exactly how the neurobiological changes produced by early
These processes occur within the olfactory bulb. experiences might persistently alter the neurobiological
In mothers and offspring, NE system activation increases mechanisms providing for later maternal learning and plas-
the signal-to-noise ratio for odors that are present, and ticity constitutes the next challenge.
promotes consolidation of neuronal changes at the cellular
and synaptic level in the olfactory bulb. Further, the initial
Acknowledgements
discrimination and its response seems to prime or sensitize
olfactory-affective systems for more rapid or efficient subse-
Many thanks to Bernie Schiff, David Sherry, Sarah Shet-
quent discriminations and associations. Finally, in both the
tleworth, and Jeff Alberts and his students for their very
mother and the offspring, neuropeptides (oxytocin and
helpful comments on earlier versions of this manuscript.
endogenous opioids) were shown to enhance affiliative
Research and manuscript preparation supported by
behaviors that are dependent on olfactory and somatosen-
M.R.C., N.S.E.R.C. and S.S.H.R.C., to A.S. Fleming, The
sory input. Collectively, these findings suggest that comple-
John D. and Catherine T. MacArthur Foundation, NIMH
mentary mechanisms mediate mother-infant affiliation in
(MH 40748), NICHHD (HD 23042), NIAAA (AA 10079)
mammals. The association of maternal odors with beha-
to G.W. Kraemer and, N.S.E.R.C. to D.H. O’Day.
vioral affiliation responses in infants may well prepare the
developing nervous system for rapid expression of maternal
behavior in response to infant odors in adulthood. Stated References
another way, the development of the rat pup’s ability to
discriminate maternal odor means that the neural machinery [1] Rosenblatt JS, Snowdon C. Parental care: evolution, mechanisms,
from olfactory receptor mechanisms to more central proces- and adaptive significance. Advances in the study of behavior, 25.
New York: Academic Press, 1996.
sing was altered. This is likely to determine to some extent [2] Alberts JR, Gubernick DJ. Functional organization of dyadic and
how pup odors are processed and discriminated when the tryadic parent–offspring systems. In: Krasnegor NA, Bridges RS,
pup becomes a mother. editors. Mammalian parenting, New York: Oxford University
Mothers also depend on postpartum experiences with Press, 1990 p. 416–440.
their young to sustain their responsiveness during lactation, [3] Blass E. Ontogeny of ingestive behavior. In: Fluharty S, Morrison
AR, Sprague JM, Stellar E, editors. Progress in psychobiology and
i.e. well past the period of hormonal priming. Experiences physiological psychology, 16. San Diego: Academic Press, 1995
with offspring earlier in life or after a previous birth also p. 1–5.
enhance, refine or tune maternal responsiveness towards [4] Horn G. Neural basis of recognitioni memory investigated through
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 683

an analysis of imprinting. Phil Trans Royal Soc Lond Series B cells in the rat main olfactory bulb. Neuroscience 1996;72:1073–
1990;329:133–142. 1082.
[5] Lorenz K. Der kumpan in der umwelt des vogels. Journal fur [26] Yu GZ, Kaba H, Okutani F, et al. The olfactory bulb: a critical site of
Ornithologie 1970;83:137–213 see also p. 289–413; Reprinted. In: action for oxytocin in the induction of maternal behavior in the rat.
Martin R, editor. Studies in animal and human behavior, vol. 1. Neuroscience 1996;72:1083–1088.
London: Meuthuen, 1970. p. 101–258. [27] Leon M, Coopersmith R, Beasley L, Sullivan RM. Thermal aspects
[6] Nordeen KW. Neural correlates of sensitive periods in avian song of parenting. In: Krasnegor NA, Bridges RS, editors. Mammalian
learning. Ann NY Acad Sci 1997;807:386–400. parenting: biochemical, neurobiological, and behavioral determi-
[7] Sherry DF, Duff SJ. Behavioural and neural bases orientation in nants, New York: Oxford University Press, 1990 p. 400–415.
food-storing birds. J Expt Biol 1996;199:165–171. [28] Lee A, Watchus J, Fleming AS. Neuroanatomical basis of maternal
[8] Shettleworth SJ. Varieties of learning and memory in animals. J Exp memory in postpartum rats: selective role for the nucleus accum-
Psychol: Anim Behav Processes 1993;19:5–14. bens. Behavioral Neuroscience 1999; in press.
[9] Corter C, Fleming AS. Psychobiology of maternal behavior in [29] Fleming AS, Morgan HD, Walsh C. Experiential factors in postpar-
human beings. In: Bornstein M, editor. Handbook of parenting, 2. tum regulation of maternal care. In: Rosenblatt JS, Snowdon CT,
New Jersey: L Erlbaum Associates, 1995:87–116. editors. Advances in the study of behavior, 25. San Diego: Academic
[10] Pryce CR. Socialization, hormones, and the regulation of maternal Press, 1996 p. 295–332.
behavior in non-human simian primates. In: Rosenblatt JS, Snowdon [30] Rosenblatt JS. Nonhormonal basis of maternal behavior in the rat.
CT, editors. Advances in the study of behavior, 25. New York: Science 1967;156:1512–1514.
Academic Press, 1996 p. 423–473. [31] Fleming AS, Cheung US, Myhal N, Kessler Z. Effects of maternal
[11] Gomendio M. Maternal styles in old world primates: their adaptive hormones on ‘timidity’ and attraction to pup-related odors in female
significance. In: Pryce CR, Martin RD, Skuse D, editors. Mother- rats. Physiol Behav 1989;46:449–453.
hood in human and nonhuman primates: biosocial determinants, [32] Fleming AS, Vaccarino F, Leubke C. Amygdaloid inhibition of
Switzerland: Karger, Basel, 1995 p. 59–68. maternal behavior in the nulliparous female rat. Physiol Behav
[12] Holman SD, Goy RW. Experiential and hormonal correlates of care- 1980;25:731–743.
giving in rhesus macaques. In: Pryce CR, Martin RD, Skuse D, [33] Fleming AS, Sarker J. Experience-hormone interactions and mater-
editors. Motherhood in human and nonhuman primates: biosocial nal behavior in rats. Physiol Behav 1990;47:1165–1173.
determinants, Switzerland: Karger, Basel, 1995 p. 87–93. [34] Fleming AS, Korsmit M, Deller M. Rat pups are potent reinforcers to
[13] Maestripieri D, Wallen K, Carrol KA. Infant abuse runs in families the maternal animal: effects of experience, parity, hormones and
of group-living pigtail macaques. Child Abuse Neglect dopamine function. Psychobiol 1994;22:44–55.
1997;21:465–471. [35] Lee AW, Fleming AS, Clancy S, Smith ML. Maternal motivation:
[14] Fairbanks LA. Individual differences in maternal style: causes and the effect of medial preoptic lesions on operant behavior in female
consequences for mothers and offspring. In: Rosenblatt JS, Snowdon rats. Soc Neuro 1996;22:454–455.
CT, editors. Advances in the study of behavior, San Diego: [36] Orpen BB, Fleming AS. Experience with pups sustains maternal
Academic Press, 1996 p. 579–611. responding in postpartum rats. Physiol Behav 1987;40:47–54.
[15] Poindron P, Levy F. Physiological, sensory, and experimental deter- [37] Morgan HD, Fleming AS, Stern JM. Somatosensory control of the
minents of maternal behavior in sheep. In: Krasnegor NA, Bridges onset and retention of maternal responsiveness in primiparous
RS, editors. Mammalian parenting: biochemical, neurobiological Sprague-Dawley rats. Physiol Behav 1992;51:541–555.
and behavioral determinants, New York: Oxford University Press, [38] Stern JS. Somatosensation and maternal care in Norway rats. In:
1990 p. 133–157. Rosenblatt JS, Snowdon CT, editors. Advances in the study of beha-
[16] Formby D. Maternal recognition of infant’s cry. Dev Med Child vior, San Diego: Academic Press, 1996 p. 243–294.
Neuro 1967;9:292–298. [39] Magnusson JE, Fleming AS. Rat pups are reinforcing to the maternal
[17] Kaitz M, Lapidota P, Bonner R, Eidelman A. Parturient women can rat: role of sensory cues. Psychobiology 1995;23(1):69–75.
recognize their infants by touch. Dev Psychol 1992;28:35–39. [40] Fleming AS, Kucera C, Lee A, Winocur G. Olfactory-based social
[18] Schaal B, Porter RH. Microsmatic humans revisited: the generation learning varies as a function of parity in female rats. Psychobiol
and perception of chemical signals. In: Slater PJB, Rossenblatt JS, 1994;22:37–43.
Milinski M, editors. Advances in the study of behavior, San Diego: [41] Fleming AS, Cheung US, Barry M. Cycloheximide blocks the reten-
Academic Press, 1991 p. 135–199. tion of maternal experience in postpartum rats. Behav Neur Biol
[19] Carter CS, Lederhendler II, Kirkpatrick B. The integrative neuro- 1993;53:64–73.
biology of affiliation. Ann NY Acad Sci 1997;87:00. [42] Moffat SD, Suh EJ, Fleming AS. Noradrenergic involvement in the
[20] Krasnegor NA, Bridges RS. Mammalian parenting: biochemical, consolidation of maternal experience in postpartum rats. Physiol
neurobiological and behavioral determinants. New York: Oxford Behav 1993;53:805–811.
University Press, 1990. [43] Numan M, Sheehan TP. Neuroanatomical circuitry for mammalian
[21] Bridges RS. Biochemical basis of parental behavior in the rat. In: maternal behavior. Ann NY Acad Sci 1997;807:101–125.
Rosenblatt JS, Snowdon CT, editors. Advances in the study of beha- [44] Modney BK, Hatton GI. Motherhood modifies magnocellular neuro-
vior, 25. San Diego: Academic Press, 1996 p. 215–242. nal relationships in functionally meaningful ways. In: Krasnegor
[22] Levy F, Kendrick KM, Keverne EB, Proter RH, Romeyer A. Physio- NA, Bridges RS, editors. Mammalian parenting: biochemical,
logical, sensory, and experiential factors of prenatal care in sheep. neurobiological, and behavioral determinants, New York: Oxford
In: Rosenblatt JS, Snowdon CT, editors. Advances in the study of University Press, 1990 p. 305–323.
behavior, 25. San Diego: Academic Press, 1996 p. 385–416. [45] Modney BK, Yang QZ, Hatton GI. Pup-induced maternal behavior
[23] Kendrick KM, Guevara-Guzman R, Zorrilla J, et al. Formation of in virgin rats is associated with increased frequency of dye coupling
olfactory memories mediated by nitric oxide. Nature 1997;388:670– among supraoptic nucleus (SON) neurons. Soc Neurosci Abstr
674. 1987;13:1593.
[24] Kendrick KM, Levy F, Keverne EB. Changes in the sensory proces- [46] Grosvenor CE, Shah GV, Crowley WR. Role of neurogenic stimuli
sing of olfactory signals induced by birth in sheep. Science and milk production in the regulation of prolactin secretion during
1992;256:833–836. lactation. In: Krasnegor NA, Bridges RS, editors. Mammalian
[25] Yu GZ, Kaba H, Okutani F, et al. The action of oxytocin originating parenting: biochemical, neurobiological, and behavioral determi-
in the hypothalamic paraventricular nucleus on mitral and granule nants, New York: Oxford University Press, 1990 p. 324–342.
684 A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685

[47] Sagar SM, Sharp FR, Curran T. Expression of c-fos protein in the [69] Suzuki T. Protein kinases involved in the expression of long-term
brain: Metabolic mapping at the cellular level. Science potentiation. Int J Biochem 1994;26:735–744.
1988;240:132–133. [70] Shibasaki F, Price ER, Milan D, et al. Role of kinases and the
[48] Jackson SP. Regulating transcription factor activity by phosphoryla- phosphatase calcineurin in the nuclear shuttling of transcription
tion. Trends Cell Biol 1992;2:104–108. factor NF-AT4. Nature 1996;382:370–373.
[49] Calamandrei G, Keverne EG. Differential expression of Fos protein [71] Haruta T, Takami N, Ohmura M, et al. Ca 21-dependent interaction
in the brain in the brain of female mice dependent on pup sensory of the growth-associated protein GAP-43 with the synaptic core
cues and maternal retrieving by rats. Behav Neurosci 1994;108:113– complex. Biochem J 1997;325:455–463.
120. [72] Lydan MA, O’Day DH. Production of 35S-labeled proteins in E. coli
[50] Fleming AS, Korsmit M. Plasticity in the maternal circuit: effects of and their use as molecular probes. Meth Mol Biol 1994;31:389–396.
maternal experience on Fos-lir in hypothalamic, limbic and cortical [73] Xerri C, Stern JM, Merzenich M. Alterations of the cortical repre-
structures in the postpartum rat. Behav Neuro 1996;110(3):567–582. sentation of the rat ventrum induced by nursing. J Neurosci
[51] Fleming AS, Suh EJ, Korsmit M, Rusak B. Activation of fos-like 1994;14:1710–1721.
immunoreactivity in MPOA and limbic structures by maternal and [74] Romanski L, Clugnet M, Bordi F, Le Doux JE. Somatosensory and
social interactions in rats. Behav Neuro 1994;108:724–734. auditory convergence in the lateral nucleus of the amygdala. Behav
[52] Walsh C, Fleming AS, Lee A, Magnusson J. The effects of olfactory Neurobiol 1993;107:444–450.
and somatosensory desensitization on fos-like immunoreactivity in [75] Vaccarino FJ, Schiff BB, Glickman SE. A biological view of rein-
the brains of pup-exposed postpartum rats. Behav Neuro forcement. In: Klein SB, Mowrer RR, editors. Contemporary learn-
1996;110(1):1–20. ing theories, Mahwah, New Jersey: L Erlbaum Associates, 1989 p.
[53] Brown JR, Ye H, Bronson RT, Dikkes P, Greenberg ME. A defect in 111–142.
nurturing in mice lacking the immediate early gene fosB. Cell [76] Pederson C, Blass E. Olfactory control over suckling in albino rats.
1996;86:297–309. In: Aslin RN, Alberts JR, Petersen MR, editors. The development of
[54] Numan M, Numan MJ. Expression of fos-like immunoreactivity in perception: psychobiological perspectives, New York: Academic
the preoptic region of maternally behaving virgin and postpartum Press, 1981 p. 359–381.
rats. Behav Neurobiol 1994;108:379–394. [77] Rosenblatt JS. Suckling and home orientation in the kitten: a
[55] Morgan HD, Watchus JA, Fleming AS. The effects of electrical comparative developmental study. In: Tobach E, Aronson L, Shaw
stimulation of the medial preoptic area and the medial amygdala E, editors. The biopsychology of development, New York:
on maternal responsiveness in female rats. Ann NY Acad Sci Academic Press, 1971 p. 345–410.
1997;807:602–605. [78] Galef Jr BG. An adaptationist perspective on social learning, social
[56] Everitt B, Robbins T. Amygdala-ventral straital interactions and feeding, and social foraging in Norway rats. In: Dewsbury DA,
reward-related processes. In: Aggleton J, editor. The amygdala: editor. Contemporary issues in comparative psychology, Suderland,
neurobiological aspects of emotion, memory, and dysfunction, MA: Sinauer, 1990 p. 55–79.
New York: Wiley-Liss, 1992 p. 401–430. [79] Harlow HF. The maternal affectional system of rhesus monkeys. In:
[57] Gaffan D. Amygdala and the memory of reward. In: Aggleton J, Rheingold HL, editor. Maternal behavior in mammals, New York:
editor. The amygdala: neurobiological aspects of emotion, memory, John Wiley and Sons, 1963 p. 254–281.
and dysfunction, New York: Wiley-Liss, 1992 p. 471–483. [80] Leon ML. Filial responsiveness to olfactory cues in the laboratory
[58] White NA. Functional hypothesis concerning the striatal matrix rat. In: Lehrman DS, Hinde RA, Shaw E, editors. Advances in the
patches mediation of S-R memory and reward. Life Sciences study of behavior, New York: Academic Press, 1978 p. 117–153.
1989;43:1943–1957. [81] Malenfant SA, Barry M, Fleming AS. Effects of cycloheximide on
[59] Stein B, Meredith M. The merging of the senses. Cambridge: MIT the retention of olfactory learning and maternal experience effects in
press, 1993. postpartum rats. Physiol Behavior 1991;49:289–294.
[60] Ghosh A, Greenberg ME. Calcium signaling in neurons: molecular [82] Rozin P, Schull RC. The adaptive-evolutionary point of view in
mechanisms and cellular consequences. Science 1995;268:239–247. experimental psychology. In: Atkinson RC, Herrnstein RJ, Lindzey
[61] Albarracin CT, Palfrey HC, Duan WR, et al. Prolactin regulation of G, Luce RD, editors. Steven’s handbook of experimental psychol-
the calmodulin-dependent protein kinase III elongation factor-2 ogy, 1. New York: Wiley, 1988 p. 503–546, 2nd edition.
system in the rat corpus luteum. J Biol Chem 1994;269:7772–7776. [83] Hall WG. The ontogeny on ingestive behavior: changing control of
[62] Newton AC. Regulation of protein kinase C. Curr Opin Cell Biol components in the feeding sequence. In: Striker EM, editor. Hand-
1997;9:161–167. book of behavioral neurobiology: neurobiology of food and fluid
[63] Nishizuka Y. Protein Kinase C and lipid signalling for sustained intake, 10. New York: Plenum Press, 1990 p. 77–123.
cellular responses. J FASEB 1995;9(7):484–496. [84] Moore C. Maternal contributions to mammalian reproductive devel-
[64] Huang YY, Colley PA, Routtenberg A. Postsynaptic the presynaptic opment and the divergence of males and females. In: Slater PJB,
protein kinase C activity may be necessary for long-term potentia- Rosenblatt JS, Snowdon CT, Milinski M, editors. Advances in the
tion. Neurosci 1992;49:819–827. study of behavior, 24. San Diego: Academic Press, 1995 p. 47–118.
[65] Olds JL, Alkon DL. Protein kinase C: a nexus in the biochemical [85] Panksepp J, Nelson E, Bekkedal M. Brain systems for the mediation
events that underlie associative learning. Acta Neurobiol Exp of social separation—distress and social-reward. Evolutionary ante-
1993;53:197–207. cedents and neuropeptide intermediaries. Ann NY Acad Sci
[66] Ambalavanar R, VanderZee E, Bolhuis J, McCabe B, Horn G. Co- 1997;807:78–100.
expression of Fos immunoreactivity in PKC-positive neurons: quan- [86] Wilson DA, Sullivan RM. Review: neurobiology of associative
titative analysis of a brain region involved in learning. Brain Res learning in the neonate: Early olfactory learning. Behav Neural
1993;606:315–318. Biol 1994;61:1–18.
[67] Fleming AS, Featherstone R, Watchus J, Smyth R, Lydan M, O’Day [87] Najbauer J, Leon M. Olfactory experience mediates apoptosis in the
D, Ivy G. Neuroplasticity of the maternal brain: changes in signal developing olfactory bulb. Brain Res 1995;674:245–251.
transduction and glial proteins associated with postpartum experi- [88] Nelson E, Alberts RA. Oxytocin-induced paw suckling in infant rats.
ence in rats. Soc Neurosci 1996;x:22. Ann NY Acad Sci 1997;807:543–548.
[68] Sheng M, Thompson MA, Greenberg ME. CREB: a Ca 21-regulated [89] Liu D, Diorio J, Tannenbaum B, et al. Maternal care, hippocampal
transcription factor phosphorylated by calmodulin-dependent glucocorticoid receptors, and hypothalamic-pituitary-adrenal
kinases. Science 1991;252:1427–1430. responses to stress. Science 1997;277:1659–1662.
A.S. Fleming et al. / Neuroscience and Biobehavioral Reviews 23 (1999) 673–685 685

[90] Francis D, Diorio J, LaPlante P. The role of early environmental [98] Suomi SJ, Ripp C. A history of motherless mothering at the Univer-
events in regulating neuroendocrine development. Ann NY Acad sity of Wisconsin Primate Laboratory. In: Reite M, Caine N, editors.
Sci 1996;794:136–152. Child abuse: The non-human primate data, New York: Alan R. Liss,
[91] Kalin NH, Shelton SE, Takahashi LK. Defensive behaviors in rhesus 1983 p. 49–78.
monkeys: Ontogeny and context-dependent selective expression. [99] Kraemer GW. A psychobiological theory of attachment. Behav
Child Dev 1991;62:1175–1183. Brain Sci 1992;15:493–511.
[92] Fonagy P, Steele H, Steele M. Maternal representations of attach- [100] Werner EE. High risk children in young adulthood: a longitudinal
ment during pregnancy predict the organization of infant–mother study from birth to 32 years. Am J Orthopsychiatry 1989;59:72–81.
attachment at one year of age. Child Dev 1991;62:891–905. [101] Seligman MEP, Hager JL, editors. Biological boundaries of learning
[93] Goldberg S, MacKay S, Rochester M. Affect attachment maternal New York: Appleton-Century Crofts, 1972.
responsiveness. Infant Behav Devel 1994;17:335–339. [102] Slater PJB. Bird song learning: causes and consequences. Ethology
[94] Waters E, Merrick SK, Albersheim LJ, Treboux A. Attachment Ecol Evol 1989;1:19–46.
security from infancy to early adulthood: a 20 year longitudinal [103] Carlson M, Earls F. Psychological and neuroendocrinological seque-
study of attachment security in infancy and early adulthood. Meeting lae of early social deprivation in institutionalized children in Roma-
presentation Society for Research on Child Development (SRCD), nia. Ann NY Acad Sci 1997;807:419–428.
Indianapolis, Indiana, USA, 1995. [104] Chiccetti D, Carlson V. Child maltreatment: theory and research on
[95] Clarke AS, Schneider ML. Prenatal stress has long-term effects on the causes and consequences of child abuse and neglect. New York:
behavioral responses to stress in juvenile rhesus monkeys. Dev Cambridge University Press, 1989.
Psychobiol 1993;26:293–304. [105] Kaufman J, Zigler E. Child maltreatment: theory and research on the
[96] Kraemer GW. Attachment and Psychopathology. Behav Brain Sci causes and consequences of child abuse and neglect. New York:
1992;15:512–541. Cambridge University Press, 1989 p. 129–150.
[97] Kraemer GW, Clarke AS. Social attachment, brain function, and [106] Pedersen C, Caldwell JD, Jirikowski GF, Insel TR, editors. Oxytocin
aggression. Ann NY Acad Sci 1997;794:121–135. in material, sexual, and social behaviors. Ann NY. Acad, Vol. 652.

You might also like