hobb: 00003 - elsevier

31
ELSEVIER THIRD PROOF 3 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain G Gonza ´ lez-Mariscal, CINVESTAV-Universidad Auto ´ noma de Tlaxcala, Tlaxcala, Mexico C H Kinsley, University of Richmond, Richmond, VA, USA ß 2009 Elsevier Inc. All rights reserved. Chapter Outline 3.1 Introduction 2 3.2 Endocrine Control 2 3.2.1 Prepartum Activities 2 3.2.1.1 Nest-building 2 3.2.1.2 Maternal aggression and isolation behavior 3 3.2.1.3 Food intake 3 3.2.2 Behaviors at Parturition 3 3.2.3 Postpartum Activities 4 3.2.3.1 Nursing 4 3.2.3.2 Maternal behavior without nursing 5 3.3 Neural Control 5 3.3.1 Cortex, Trigeminal Complex, and Olfactory Circuit 5 3.3.2 Septum and Bed Nucleus of the Stria Terminalis 6 3.3.3 MPOA and Its Connections 7 3.3.3.1 Studies using lesion and deafferentation techniques 7 3.3.3.2 Studies using implantation of hormones and detection of their receptors 8 3.3.4 Midbrain Tegmentum, Paraventricular Nucleus, and Habenular Complex 9 3.3.5 Sampling the Active Parental Brain 10 3.3.5.1 The expression of immediate early genes 10 3.3.5.2 Other methods 12 3.4 Epigenetic, Intergenerational Transmission of Mothering Styles: Its Impact on Offspring Development 12 3.5 Impact of Maternal Behavior on the Mother: Effects on Cognition and Neuroplasticity 15 3.5.1 Theoretical Context 15 3.5.2 Fear and Anxiety Regulation 16 3.5.2.1 Behaviors affected 16 3.5.2.2 Brain mechanisms 16 3.5.3 Regulation of Cognitive Effects 17 3.5.3.1 Learning and memory 17 3.5.3.2 Neural regulation of cognitive effects 17 3.6 Conclusions and Perspectives 19 References 20 Further Reading 28 Glossary g0005 dendritic spines Tiny protuberances arising from the pinching-off and extension of the dendrite of neurons, which are sensitive to presynaptic stimulation, neurotrophic factors, and selected hormones. Such spines provide substantial increases to the surface area of an individual neuron, and therefore differential changes to its information processing capacity. g0010 maternal behavior A constellation of behaviors that is designed to ensure the survival and 1 HOBB: 00003

Upload: others

Post on 03-Dec-2021

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

3 From Indifference to Ardor: The Onset, Maintenance,and Meaning of the Maternal BrainG Gonzalez-Mariscal, CINVESTAV-Universidad Autonoma de Tlaxcala, Tlaxcala, Mexico

C H Kinsley, University of Richmond, Richmond, VA, USA

� 2009 Elsevier Inc. All rights reserved.

Chapter Outline

3.1 Introduction 2

3.2 Endocrine Control 2

3.2.1 Prepartum Activities 2

3.2.1.1 Nest-building 2

3.2.1.2 Maternal aggression and isolation behavior 3

3.2.1.3 Food intake 3

3.2.2 Behaviors at Parturition 3

3.2.3 Postpartum Activities 4

3.2.3.1 Nursing 4

3.2.3.2 Maternal behavior without nursing 5

3.3 Neural Control 5

3.3.1 Cortex, Trigeminal Complex, and Olfactory Circuit 5

3.3.2 Septum and Bed Nucleus of the Stria Terminalis 6

3.3.3 MPOA and Its Connections 7

3.3.3.1 Studies using lesion and deafferentation techniques 7

3.3.3.2 Studies using implantation of hormones and detection of their receptors 8

3.3.4 Midbrain Tegmentum, Paraventricular Nucleus, and Habenular Complex 9

3.3.5 Sampling the Active Parental Brain 10

3.3.5.1 The expression of immediate early genes 10

3.3.5.2 Other methods 12

3.4 Epigenetic, Intergenerational Transmission of Mothering Styles:

Its Impact on Offspring Development 12

3.5 Impact of Maternal Behavior on the Mother: Effects on Cognition and

Neuroplasticity 15

3.5.1 Theoretical Context 15

3.5.2 Fear and Anxiety Regulation 16

3.5.2.1 Behaviors affected 16

3.5.2.2 Brain mechanisms 16

3.5.3 Regulation of Cognitive Effects 17

3.5.3.1 Learning and memory 17

3.5.3.2 Neural regulation of cognitive effects 17

3.6 Conclusions and Perspectives 19

References 20

Further Reading 28

Glossaryg0005 dendritic spines Tiny protuberances arising from

the pinching-off and extension of the

dendrite of neurons, which are sensitive

to presynaptic stimulation, neurotrophic

factors, and selected hormones. Such

spines provide substantial increases to

the surface area of an individual neuron,

and therefore differential changes to its

information processing capacity.

g0010maternal behavior A constellation of behaviors

that is designed to ensure the survival and

1

HOBB: 00003

Page 2: HOBB: 00003 - Elsevier
Page 3: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

These results coincide with the observation that therabbit MPOA has a high concentration of receptorsfor estrogens (ERa; Caba et al., 2003a) and for pro-gestins (PRs; Caba et al., 2003b). In sows receptorsfor PGF2a have been reported in the brain (Burne,2000). Surprisingly, few studies have explored thehormonal control of nest-building in rodents. In rats(Denenberg et al., 1969), mice (Lisk, 1971), and ham-sters (Swanson and Campbell, 1979), some combi-nations of estradiol and progesterone promote theaccumulation of shredded material but, to our knowl-edge, the brain sites where these hormones act topromote nest-building are unknown.

s0025 3.2.1.2 Maternal aggression and isolation

behaviorp0015 Several rodents show aggression to intruders in late

pregnancy; estradiol and progesterone are some ofthe factors involved in stimulating this activity (forreview, see Numan et al. (2006)), which is maximalacross lactation (see below). Shortly before parturi-tion ungulates (i.e., sheep, goats, and sows) isolatethemselves from the herd, show restlessness and agi-tation, and they emit vocalizations (Poindron et al.,2007a,b; Algers and Uvnas-Moberg, 2007). Althoughthe participation of hormones in these activities hasnot been directly explored, their occurrence close toparturition suggests an involvement of the hormonesthat time and control delivery.

s0030 3.2.1.3 Food intakep0020 Changes in this parameter have been reported during

the peripartum period in some species, both in termsof quantity (food intake declines by about 60% onprepartum day 1 in rabbits; Gonzalez-Mariscal et al.,1994) and quality (meaty foodstuffs are consumed byrodents (Gregg and Wynne-Edwards, 2006; Kristal,1991) and rabbits (Melo and Gonzalez-Mariscal,2003); amniotic fluid is consumed by ungulates(Poindron et al., 2007a)). There is evidence that and-rogens decrease food intake in rabbits (Gonzalez-Mariscal et al., 2003) but hardly anything is knownabout the hormonal factors modulating food prefer-ences around delivery in any species.

s0035 3.2.2 Behaviors at Parturition

p0025 Placentophagia and amniotic fluid ingestion are con-sistently observed following delivery in most mam-mals studied (Poindron et al., 2007a; Gregg andWynne-Edwards, 2006; Kristal, 1991; Melo andGonzalez-Mariscal, 2003). Little is known about the

consequences of placentophagia on the mother,although an analgesic effect has been shown in rats(Kristal, 1991); for the newborn the licking theyreceive from the mother as she consumes the amni-otic fluid that bathes them stimulates their breathing,dries their fur, and promotes later learning abilities.The hormonal control of placentophagia is unknown,but likely involves steroid hormones.

p0030The interaction between mother and offspring atparturition is critical to ensure the establishment ofmaternal responsiveness across lactation, especiallyin primiparous females. Clear evidence in rabbits(Gonzalez-Mariscal et al., 1998), rats (Bridges, 1975),sheep (Poindron et al., 2007a), and goats (Poindronet al., 2007b) has shown that interfering with mother/young contact at this time disrupts or seriously altersmaternal behavior. A similar interference in earlylactation provokes milder effects. These resultssuggest that the particular conditions that exist atparturition – in terms of changes in the concentrationof several hormones in blood (e.g., oxytocin (OT);PGF2a; corticosteroids; PRL; progesterone; estra-diol; for review, see Gonzalez-Mariscal and Poindron(2002)), contractions of the uterus, changes in thedistribution, and/or number of receptors for specifichormones and neurotransmitters in the brain, simul-taneously with the multisensorial perception of sti-muli from the newborn – favor the consolidation anddirect the responsiveness of the mother to the young.In the case of ungulates an additional process takesplace at this time: the recognition of the mother’s ownoffspring which determines the exclusive nursing of aparticular lamb or kid across lactation (see the follow-ing section). Although parturition has been longrecognized as a critical period for the consolidationof maternal behavior, little is known about the factorsunderlying this process. Yet, some results obtained inrats suggest that a pathway similar to learning oper-ates at this time. Thus: (1) there is direct relationshipbetween the duration of the maternal experience andthe duration of its retention following separationfrom the litter (Bridges, 1977); (2) drugs that blockprotein synthesis and interfere with several formsof learning disrupt maternal behavior (Fleminget al., 1996), especially when placed directly into theshell of the nucleus accumbens (Li and Fleming,2003); (3) removing corticosteroids, which are knownto favor the consolidation of some forms of learning,prior to parturition (by adrenalectomy in late preg-nancy) reduces maternal responsiveness followingseparation from the litter for several days (Grahamet al., 2006).

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 3

HOBB: 00003

Page 4: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

s0040 3.2.3 Postpartum Activities

s0045 3.2.3.1 Nursingp0035 This is undoubtedly the main behavior displayed by

all mammalian mothers toward their progeny fromparturition until weaning. Yet, the specific character-istics of nursing and the display of activities thatsurround it vary greatly among species. For instance,in rodents, nursing is preceded by retrieving the pupsthat have strayed away and grouping them in thenest. Rabbits and sows neither retrieve their young(despite their small size) nor do ewes or goats (whoproduce large, well-developed offspring). The pre-dictable, unfailing display of nursing and its asso-ciated behaviors depends on several factors thatact from late pregnancy and throughout lactation.Among the former, PRL released before parturitionin rats (Andrews and Grattan, 2002) and rabbits(Gonzalez-Mariscal et al., 2000) plays a criticalrole in promoting maternal responsiveness: prevent-ing PRL secretion (by intracerebroventricular (ICV)injection of bromocryptine) in the last 5 days ofpregnancy prevents the display of nursing from par-turition onward in rabbits (Gonzalez-Mariscal et al.,2004). This result agrees with the observation that anincrease in the density of the long form of the PRLreceptor occurs in specific brain regions (notably, theMPOA) at the end of pregnancy in rats (Bakowskaand Morrell, 1997).

p0040 At parturition, vaginocervical stimulation (VCS)plays a major role in the establishment of maternalbehavior in ewes and goats: peridural anesthesiaresults in the failure to accept the suckling attemptsof the newborn in primiparous sheep and goats(Poindron et al., 2007a,b). Moreover, cesarean sectionprovokes a similar effect in sheep and rats. Experi-mental evidence suggests that these effects may bemediated by the release of OT occurring at parturi-tion: in rats, ICV injections of an OTantagonist at thistime prevent the onset of maternal behavior (VanLeengoed et al., 1987), and in sheep, the disruptiveeffect of peridural anesthesia is partly overcome byICV injection of OT (Krehbiel et al., 1987). Similarresults have been obtained in nonpregnant animals:VCS (Kendrick et al., 1991) or ICV OT injections(Kendrick et al., 1987) promote maternal behavior inmultiparous ewes primed with ovarian steroids; con-versely, an OT antiserum reduces the facilitation ofmaternal behavior normally seen in virgin rats primedwith ovarian steroids (Pedersen et al., 1985). Theabove evidence supports the idea that the concur-rence of specific somatosensory and hormonal factors

at parturition is critical for the onset of maternalresponsiveness across lactation. In contrast, the obser-vation that neither OT antagonist in rats (Fahrbachet al., 1985) nor bromocryptine in rats (Stern, 1977) orrabbits (Gonzalez-Mariscal et al., 2000) antagonizematernal behavior when given in early lactation sug-gests that the maintenance of responsiveness to theyoung does not involve OTor PRL.

p0045The fine-tuning of maternal behavior, that is, thedisplay of nursing with its associated species-typicalethogram throughout lactation, depends largely onsomatosensory factors. Thus, in species that givebirth to multiple young (like rodents, rabbits, andpigs), recognition of the mother’s own progeny isirrelevant as they effectively nurse young otherthan their own (Algers and Uvnas-Moberg, 2007;Gonzalez-Mariscal and Gallegos, 2007; Rosenblattand Lehrman, 1963). In these animals stimuli comingfrom the progeny regulate other aspects of maternalbehavior. Specifically, in rats the adoption of a high-crouch nursing posture critically depends on thequantity and quality of the ventral stimulation pro-vided by the suckling pups (Stern and Johnson, 1990;Stern et al., 1992). Moreover, in rats the number andduration of nursing bouts per day declines from mid-lactation onward (as does the amount ofmilk produced)in relation to the intensity of suckling, which becomesmore vigorous as the pups grow (Mena andGrosvenor,1972). Furthermore, suckling stimulation is critical formaintaining thehigh levels of aggression seen inmotherrats across lactation: pup removal leads to a decline inthis behavior within 24 h (Erskine et al., 1978).

p0050In rabbits, nipple stimulation plays a major role inregulating the duration of nursing: the normalc. 3 min day–1 (Gonzalez-Mariscal et al., 1994;Zarrow et al., 1965) requires a minimum litter sizeof three. When a single pup is provided, the timeinside the nest box (during which nursing occurs) isgreatly increased. A similar effect is observed whenthe mother’s nipples are covered or removed (thelect-omy) or when the pups’ mouths are covered (Gonza-lez-Mariscal, 2007). In sows, suckling stimulation fromthe piglets modulates a specific component of thematernal ethogram: the mother’s grunting (Algersand Uvnas-Moberg, 2007). As the sow lies down toinitiate nursing, she emits a type of grunting thatsignals the piglets to her teats. Their suckling pro-motes the initial release of OTwhich, in turn, triggersa change in the type of maternal grunting: the pigletsrespond to it by suckling more vigorously, thus initiat-ing milk letdown.

4 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 5: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

p0055 In contrast to the above species, ewes and goatsshow selective nursing throughout lactation, that is,they will accept at the udder only their own lamb orkid and actively reject alien ones (Poindron et al.,2007a,b). The recognition of the mother’s own off-spring in these species depends on the perceptionof the olfactory signature of the newborn at parturi-tion and its consolidation as an olfactory memory(Levy et al., 2004). As lactation progresses, visualand acoustic cues begin to play an important rolein the mother’s recognition of her progeny andalso in the reciprocal process, that is, in the capa-city of the young to recognize their own mother(Poindron et al., 2007a,b; Nowak et al., 2007). Thismutual recognition is essential to adjust the physiol-ogy and behavior of both parties to each otheracross lactation.

s0050 3.2.3.2 Maternal behavior without nursingp0060 Although the hormones of pregnancy prime the

maternal brain to effectively respond to the stimulishe receives from the young starting at parturition,nonpregnant, nonlactating rodents and rabbits canbehave maternally under specific conditions. Thus,rats, hamsters, gerbils, and laboratory (but not wild)mice gradually behave maternally to foster youngafter several days of cohabitation with them (for areview, see Gonzalez-Mariscal and Poindron (2002)).This so-called sensitization process does not occurin intact rabbits (Gonzalez-Mariscal et al., 2004). Yet,in both rats (Fleming and Rosenblatt, 1974a,b) andrabbits (Gonzalez-Mariscal et al., 2004; Chirino et al.,2007), lesions to the main olfactory system (MOS) orthe accessory olfactory system (AOS) stimulate thedisplay of maternal behavior. In rats this effect isfacilitated by estrogens (reduced latency to becomematernal; Mayer and Rosenblatt, 1979), whereasin rabbits estrogens are essential (absent in OVXanimals; Gonzalez-Mariscal et al., 2004; Chirinoet al., 2007).

p0065 These results indicate that the neural circuitsunderlying the motivation to interact with the prog-eny and the regulation of species-typical behaviorsare present in all female rats and rabbits, regardless oftheir reproductive status, but are tonically inhibitedby the MOS and AOS. The way through which that

Page 6: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

proposed that this behavior is under a multisensorialcontrol because the elimination of either vision,olfaction, or tactile sensitivity of the snout and peri-oral region did not interfere with the retrieval of pups.Later work (Benuck and Rowe, 1975; Herrenkohl andRosenberg, 1972; Herrenkohl and Sachs, 1972) hasconfirmed the lack of effect of anosmia, blindness,and deafness of lactating rats on maternal behavior.More recent studies in which the perioral region wasdesensitized by either injecting a local anesthetic intothe mystacial pads or by sectioning the infraorbitalbranch of the trigeminal nerve found that these pro-cedures interfere with pup retrieval (Kenyon et al.,1981, 1983; Stern and Kolunie, 1989). The discrep-ancy between these results and those of Beach andJaynes (1956) may have been due to the fact thatBeach and Jayne allowed several weeks between thesnout desensitization and testing for maternal behav-ior, a period during which re-innervation of the peri-oral region may have occurred.

p0080 In hamsters, the total removal of the neocortexdoes not eliminate maternal behavior, but when thisprocedure is accompanied by lesions of the cingulatecortex and the underlying hippocampus, maternalbehavior is abolished (Murphy et al., 1981). In pri-mates, lesions to either the prefrontal cortex orthe anterior temporal cortex (performed at around2 months postpartum) severely disrupt the mother’sactive search for her young and provoke a loss of theprotective retrieval of the infant faced with threaten-ing situations (Bucher, 1970; Franzen and Myers,1973; Myers et al., 1973). Xerri et al. (1994) havereported that the somatosensory cortex of mothersoccupies more surface area than that of nulliparousfemales, an indication that as ever greater demandsare placed up on the mother’s responsivity to heroffspring, the brain responds by becoming more sen-sitive to their many sensory cues, including suckling,tactile, etc. This inherent neuroplasticity is an exam-ple of the expression of the female’s maternal behav-ioral state.

p0085 Lesions of the amygdala (a structure that receivesmajor input from the MOS and AOS) do not disruptmaternal behavior in lactating mice (Slotnick andNigrosh, 1975) and lesions to the stria terminalis(the major efferent pathway of the amygdala) haveno effect on the maternal care of lactating rats(Numan, 1974). By contrast, lesions of the cortico-medial amygdala (Del Cerro et al., 1991; Fleminget al., 1980; Numan et al., 1993), bed nucleus of theaccessory olfactory tract (Del Cerro et al., 1991), or

stria terminalis in virgin rats stimulate maternalbehavior (Fleming et al., 1980) while kindling-typeelectrical stimulation of the medial amygdala delaysthe onset of maternal responsiveness to foster pups inexperienced mother rats (Morgan et al., 1999).

p0090On the other hand, activity in the amygdalaappears to be related to the support of the requisitematernal behaviors a mother displays in supportof the care and maintenance of her litter. Wartellaet al. (2003) reported less activation of basolateralamygdala (BLA) in response to stressful situationsin parous (and pregnant (primi/multigravid)) com-pared to age-matched nulliparous females. Rasia-Filho et al. (2004), examining Golgi-stained neurons,reported that neuronal complexity was enhanced inthe medial amygdala in parous females. Together,these data suggest that reproductive experience mayregulate the structure and function of behavior-mediating neural substrates in ways that benefit theforaging and offspring defense capabilities of themother. A female thus equipped may be betterprepared to face the exigencies of her natural envi-ronment than the nulliparous female, whose neuralsubstrate has not yet undergone the hormonal andpup-induced changes that occur with pregnancy andits aftermath.

s00653.3.2 Septum and Bed Nucleus ofthe Stria Terminalis

p0095The septal area has been lesioned in motherrats (Fleischer and Slotnick, 1978; Terlecki andSainsbury, 1978), mice (Carlson and Thomas, 1968;Slotnick and Nigrosh, 1975), and rabbits (Cruz andBeyer, 1972). In rodents, these lesions do not abolishmaternal motivation but provoke a spatial disorgani-zation of maternal behavior: mothers pick up pups,carry them around, drop them at random (thus alter-ing crouching and nursing), and they may alsobuild several small nests where they retrieve young(Terlecki and Sainsbury, 1978). Excitotoxic aminoacid lesions of the bed nucleus of the stria termi-nalis disrupt retrieval behavior in postpartum rats(Numan, 1996). Rats lesioned in the septal regionalso show increased defensiveness, an alteration pro-posed to explain the disorganization of maternalbehavior (Sheehan et al., 2000). In contrast, thebehavior of rabbits is severely altered in all its aspectsfollowing septal lesions; mothers do not build nestsand refuse to enter the nest box for nursing (Cruz andBeyer, 1972).

6 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 7: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

s0070 3.3.3 MPOA and Its Connections

s0075 3.3.3.1 Studies using lesion and

deafferentation techniquesp0100 Much evidence has accumulated to identify the

MPOA as a critical site for the expression of maternalbehavior. Electrolytic or radiofrequency lesions of thisregion disrupt retrieving, nest-building, and nursing inlactating rats (Gray and Brooks, 1984; Jacobson et al.,1980; Numan, 1974; Numan et al., 1977) and hamsters(Marques et al., 1979; Miceli and Malsbury, 1982).A similar effect is provoked by injecting N-methyl-D-aspartate (NMDA), which selectively destroys cellbodies but spares fibers of passage, into the MPOA oflactating rats (Numan et al., 1988). Moreover, virginfemales (ovariectomized or intact) lesioned in theMPOA do not show maternal behavior despite manydays of exposure to pups (Gray and Brooks, 1984;Miceli et al., 1983; Numan et al., 1977). Furthermore,the facilitation of maternal behavior provoked invirgin rats by lesioning the corticomedial amygdaladoes not occur if such females have also beenlesioned in the MPOA (Fleming et al., 1983). Incontrast, by applying kindling-type electrical stimu-lation to the MPOA maternal responsiveness to fos-ter pups is promoted in both experienced mothersand virgins (Morgan et al., 1999), a finding support-ing the idea that activity of this brain structure iscritical for the performance of maternal behavior.Furthermore, this brain region also seems to mediatethe reinforcing properties of young pups in postpar-tum and in sensitized virgin rats: electrolytic lesionsto the MPOA disrupt maternal behavior in thehome cage and decrease the rate of bar-pressing forpups in an operant reinforcement paradigm (Leeet al., 2000).

p0105 Adifferent experimental approach has been used toinvestigate the connections of the MPOA which maybe part of a circuit regulating specific aspects of mater-nal behavior. By selectively severing the rostral, cau-dal, lateral, or dorsal connections of the MPOA andassessing the effects of such procedures on the mater-nal behavior of lactating rats, a series of experiments(Franz et al., 1986; Miceli et al., 1983; Numan, 1974;Numan and Callahan, 1980; Numan and Corodimas,1985; Terkel et al., 1979) have revealed that the lateralprojections of the MPOA exert the most importantinfluence. Knife cuts interrupting such connectionsseverely disrupt the oral components of maternalbehavior (i.e., retrieving and nest-building) and pro-voke moderate alterations in crouching and nursing.Such deficits are long-lasting, being still apparent

1 month after the knife cuts are performed ( JakubowskiandTerkel, 1986; Numan, 1990). It is important to notethat the observed deficits in pup retrieval are not aconsequence of a general inability to retrieve anything,because lesioned females readily hoard candy (Numanand Corodimas, 1985). It must also be emphasized thatthe disruption in maternal behavior that followsMPOA lesions is not an indirect consequence of altera-tions in pituitary function because: (1) this effect occurseven in hormonally primed rats (Numan andCallahan,1980); (2) such lesions disrupt the maintenance ofmaternal behavior in lactating ( Jacobson et al., 1980;Miceli et al., 1983; Numan, 1974; Numan andCallahan, 1980; Terkel et al., 1979) and in virgin(i.e., sensitized; Gray and Brooks, 1984; Miceli et al.,1983; Numan et al., 1977) rats, known to be largelyindependent of hormones.

p0110To further define the elements and pathways com-prised in a maternal circuit, Numan and colleaguesperformed a systematic series of studies involvingthe selective destruction of specific brain areas andneural connections and the determination of theireffects on the maternal behavior of lactating rats.As stated earlier, severing the lateral projections ofthe MPOA disrupts maternal behavior, an effect alsoseen after lesioning the lateral hypothalamus (Avarand Monos, 1966, 1967, 1969a,b). These lateralMPOA projections travel to the ventral tegmentalarea (VTA) by one of two routes: a direct one andan indirect one involving a synapse in the lateralpreoptic area (LPOA; Barone et al., 1981; Conradand Pfaff, 1976; Swanson, 1976). Numan has pro-posed that the VTA may be important in regulatingongoing maternal behavior because some of theascending projections of this region reach the stria-tum and are involved in the control of oral motorresponses. Indeed, bilateral electrolytic (though notNMDA) lesions of the VTA severely disrupt mater-nal behavior in postpartum rats (Gaffori and LeMoal, 1979; Numan and Smith, 1984). Bilateralknife cuts through the lateral hypothalamus disruptmaternal behavior only when performed at a level(dorsal) in which fibers coming from neurons in thelateral hypothalamus and going to the VTA are sev-ered. When cuts are made ventrally, the connectionsdisrupted are those from MPOA neurons projectingdirectly to the VTA and this procedure has no effecton maternal behavior (Numan et al., 1985). Severealterations in maternal behavior are also obtainedwhen unilateral electrolytic lesions of the VTA arecombined with contralateral knife cuts of the lateral

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 7

HOBB: 00003

Page 8: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

projections of the MPOA, thus indicating that intactbilateral connections between the MPOA and theVTA are essential for the adequate display of mater-nal behavior.

p0115 To distinguish which of the two routes connectingMPOA and VTA (i.e., the direct one or the one with arelay in the LPOA) is part of the maternal circuit,Numan et al. (1988) performed radiofrequency orNMDA lesions in the LPOA and found that bothprocedures provoked severe disruptions in maternalbehavior. This was not observed when NMDA lesionswere performed in the lateral hypothalamus. Theseresults are consistent with the finding that fiberspassing through the lateral hypothalamus (and whichare crucial for maternal behavior) come mainly fromcell bodies located in the LPOA and bed nucleus ofthe stria terminalis (Numan et al., 1985). In summary,these studies support the existence, in rats, of amaternal circuit involving the MPOA, LPOA, andafferent fibers passing through the lateral hypothala-mus and the VTA.

s0080 3.3.3.2 Studies using implantation of

hormones and detection of their receptorsp0120 Diluted estradiol was implanted into the MPOA of

rats that were ovariectomized and hysterectomizedon pregnancy day 16 (Numan et al., 1977). Thisprocedure effectively stimulated maternal behaviorand the results were confirmed in a later study(Fahrbach and Pfaff, 1986). These findings coincidewith the observation that many cells that concentrateestradiol and project to or through the ventral mid-brain are located in the MPOA, LPOA, and bednucleus of the stria terminalis, as determined by thecombined use of a fluorescent retrograde tracer(injected into the ventral midbrain) plus 3H-estradiolautoradiography (Fahrbach and Pfaff, 1986). More-over, toward the end of pregnancy there is an increasein the number of estrogen receptors in the MPOA, asdetermined by the binding of 3H-estradiol to cytosoland nuclear receptors extracted from this region(Giordano et al., 1989, 1990) and by the detection ofmRNA specific for the a-form of the ER (Wagnerand Morrell, 1996; Wagner et al., 1998).

p0125 It is unclear if progesterone acts on the MPOAto regulate maternal behavior because hormoneimplants in this region fail to inhibit maternal behav-ior induced by estradiol injections given to ovariecto-mized/hysterectomized late pregnant rats (Numan,1978). Yet, PRs are found at highest levels in latepregnancy in the MPOA and ventral part of the bednucleus of the stria terminalis, among other regions

(rat: Numan et al. (1999); guinea pig: Blausteinand Turcotte (1989), Blaustein et al. (1988), andWarembourg et al. (1986, 1989)). In sheep, the distri-bution of PRs is similar to that of other species (Scottet al., 2000), but their possible role in the control ofmaternal behavior remains unexplored.

p0130Support for an action of PRL on the MPOA tofacilitate maternal behavior comes from the find-ings of Bridges and colleagues. These investigatorsfound that implants of ovine or rat PRL, placentallactogen-I, or growth hormone into the MPOA ofovariectomized, bromocryptine-treated rats, givenprogesterone and then estradiol, readily facilitatematernal behavior (Bridges and Freemark, 1995;Bridges and Mann, 1994; Bridges et al., 1990, 1997).PRL binding sites were initially detected in thehypothalamus by the binding of 125I-ovine PRL toneuronal membranes (Muccioli and Di Carlo, 1994).Later studies have detected estrogen-induced PRLreceptors in the MPOA using immunocytochemistry(Pi and Grattan, 1998), and in situ hybridization(Pi and Grattan, 1999a). Moreover, the mRNA forthis receptor is expressed most abundantly at the endof pregnancy (Bakowska and Morrell, 1997) and dur-ing lactation (Pi and Grattan, 1999a,b,c) in severalbrain regions, including the MPOA. In rabbits, PRLbinding sites have been detected in: (1) the hypothal-amus of intact females by measuring the affinitylabeling of 125I-ovine PRL to membranes obtainedfrom the hypothalamus (Di Carlo and Muccioli,1981); (2) the MPOA and several hypothalamicnuclei (e.g., paraventricular, supraoptic, suprachias-matic, and arcuate) of intact females by autoradiogra-phy of 125I-ovine PRL (Walsh et al., 1990).

p0135The MPOA also demonstrates plasticity of a morenatural form with regard to the onset and mainte-nance of maternal behavior. Keyser-Marcus et al.(2001) reported that neurons in the MPOA becomemore complex under the hormonal stimulationof pregnancy or a pregnancy mimic. For example,whereas there was no difference between OVX anddiestrous females, both had smaller somal areas com-pared to progesterone- and estradiol-treated andlate-pregnant females. Interestingly, the area of thesoma returned to diestrus/OVX levels in lactatingfemales. Further, the authors found a significantlygreater number of dendritic branches, and signifi-cantly longer cumulative dendritic length, in preg-nant and hormone-treated groups compared to OVX,diestrous, and lactating females. The increase insomal area may indicate increased cellular activity(Miller and Erskine, 1995) and stimulatory effects on

8 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 9: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

additional neuronal structural variables may repre-sent modifications in the information-processingcapacity of this set of neurons. Therefore, pregnancyand its attendant hormonal exposure may stimulateneurons in the MPOA which then contribute (in anas-yet undetermined manner) to the display of mater-nal behavior. During the postpartum, lactationalperiod, when cues from pups primarily maintainmaternal attention, the neuronal soma appears toreturn to a prepregnancy, nonhormonally dependentstate (but still sensitive to pup cues as transducedthrough other means: see Macbeth et al. (2008), whodetected marked alterations of brain-derived neuro-trophic factor (BDNF) in parous females). These datademonstrate a striking plasticity in the brains offemales that may be reflected in modifications inpup-directed behaviors.

s0085 3.3.4 Midbrain Tegmentum,Paraventricular Nucleus, and HabenularComplex

p0140 The lateral midbrain tegmentum, which includes theperipeduncular nucleus, receives input from descend-ing preoptic efferents and also from trigeminal sen-sory pathways that pass through the VTA and carryinput from the perioral region (Nadaud et al., 1984;Smith, 1973). Lesions in this area abolish maternalaggression toward males, block milk ejection, but donot interfere with nursing behavior (Hansen andFerreira, 1986; Hansen andGummesson, 1982; Hansenand Kohler, 1984). However, knife cuts made cau-dal to the VTA (thus disrupting ascending anddescending pathways traveling through the mesen-cephalon) abolish maternal behavior (Numan andNuman, 1991). These results indicate that fiberspassing through the VTA and relevant for maternalbehavior terminate at levels lower than the mesen-cephalon (e.g., caudal periaqueductal gray (PAG)and central tegmental field). Interesting recentwork by Felicio and his colleagues has implicatedthe PAG in a form of behavioral switching, in whichthe mechanism for a decision-making apparatusappears to involve differential activation of thedorsal and ventral PAG. As the maternal femaledecides to hunt versus acting maternal, there iscoincident alteration of the activity of the PAG (asmeasured by c-fos activity) that shifts her attentionaway/toward pups/prey objects (Felicio and Canteras,2008). The data suggest that the PAG may weigh therelative merits of chasing prey (insects in the Felicioand Canteras work) versus being maternal, based

on concurrent internal states, status of the pups, envi-ronmental constraints, etc. It may play a role in decid-ing the cost:benefit ratio that follows a choice inthe mother’s day-to-day, minute-by-minute care ofher young.

p0145The paraventricular nucleus (PVN) seems to par-ticipate in the onset of maternal behavior in severalspecies through the release of OTand its parvocellularextra-hypothalamic projections. Thus, in rats: (1) OTis released in the supraoptic nucleus (SON) and thePVN at parturition (Neumann et al., 1993); (2) ultra-structural changes are observed in the SON and PVN,such as glial retraction and an increase in direct neu-ronal coupling, in the last 24 h preceding parturition(Hatton and Ellisman, 1982; Perlmutter et al., 1984;Theodosis and Poulain, 1984; Theodosis et al., 1981);(3) most of these changes disappear within 10–30 daysafter weaning (Modney and Hatton, 1990); (4) similarchanges can be observed in sensitized virgins (Salmet al., 1988) following ICV injection of OT (Theodosiset al., 1986) or in response to electrical stimulation ofthe olfactory pathways in brain slice preparations(Hatton and Yang, 1989, 1990); (5) isolation of themediobasal hypothalamus, a procedure that leavesthe PVN and its extra-hypothalamic connectionsintact but severs pathways between the PVN and themedial basal hypothalamus, abolishes milk output butdoes not affect maternal behavior (Herrenkohl andRosenberg, 1974); (6) lesions to the PVN disrupt ratmaternal behavior, although only if performed beforeits onset (Insel and Harbaugh, 1989; Numan andCorodimas, 1985); if performed after parturitionPVN lesions significantly reduce maternal aggression(Consiglio and Lucion, 1996). On the other hand,kainic acid-induced lesions of the PVN on pospartumday 2 affect retrieving behavior (but not other mater-nal behavior components (Olazabal and Ferreira,1997)), while lesions performed on day 5 postpartumwith ibotenic acid (a procedure that preferentiallylesions parvocellular neurons) or infusion of anti-sense oligonucleotides against OT increase maternalaggression (Giovenardi et al., 1997, 1998).

p0150In sheep there is strong evidence for the partici-pation of OT from the PVN in the activation ofmaternal behavior. Thus: (1) OT is released in thisstructure during parturition and retrodialysis of thispeptide induces maternal behavior in nonpregnantsteroid primed females (Da Costa et al., 1996a);(2) birth or VCS activates the PVN (as determinedby expression of the FOS protein), where oxytociner-gic neurons and fibers as well as OT receptors havebeen found (Kendrick et al., 1997); (3) the number of

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 9

HOBB: 00003

Page 10: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

OT immunoreactive neurons and mRNA transcriptsfor the OT receptor are increased in the PVN atparturition and following progesterone/estradiolpriming (Broad et al., 1993); (4) previous maternalexperience further enhances the expression of OTreceptor mRNA in the PVN (Broad et al., 1999).From this evidence, it has been proposed that atparturition OT facilitates its own release (by actingon autoreceptors), not only in the PVN itself but alsoin the brain regions it projects to, for example, MPOAand olfactory bulbs (Kendrick et al., 1997).

p0155 The lateral habenular complex is necessary for thehormonal onset of maternal behavior because cyto-toxic lesions in this region produce deficits in pupretrieval, nest-building, and nursing in rats thatwere ovariectomized/hysterectomized and injectedwith estrogen on pregnancy day 16 (Corodimaset al., 1993; Matthews-Felton et al., 1995). However,implants of estradiol in the lateral habenula fail tostimulate maternal behavior in rats that were ovari-ectomized/hysterectomized on pregnancy day 16(Matthews-Felton et al., 1999) despite the fact thatERs have been found in this region by autoradio-graphy (Pfaff and Keiner, 1973) and in situ hybridiza-tion (Wagner et al., 1998).

s0090 3.3.5 Sampling the Active Parental Brain

s0095 3.3.5.1 The expression of immediate

early genesp0160 The protein products of c-fos, fosB, egr-1, and zif-268

(examples of the so-called immediate early genes)participate in regulating the transcription of late-responding genes. A large amount of evidence sup-ports the idea that an increased production ofthe mRNA or the protein products of such genesindicates neuronal stimulation (Dacoit et al., 1990;Morgan and Curran, 1991; Wisden et al., 1990). Thus,the quantification of such gene products has beenused to measure neuronal activity in specific brainregions under a variety of experimental conditions.Because several antibodies for the FOS protein arereadily available, most studies have used the detec-tion of this protein by means of immunocytochemis-try to explore the brain regions activated during thedisplay of maternal behavior. By using this approach,Numan and Numan (1994) found that lactating ratsexposed to pups (following a 3-day separation) showedsignificantly higher numbers of FOS-immunoreactive(IR) neurons in the MPOA, ventral (though notdorsal) bed nucleus of the stria terminalis, anteriorcortical and posterodorsal medial amygdaloid nuclei,

compared to lactating rats that were given candyinstead of pups on the day of testing. Very similarchanges were observed in virgin rats behaving mater-nally after having been exposed to pups for severaldays, with respect to females also given pups for thesame number of days but not yet behaving maternally.No differences in the number of FOS-IR neuronsbetween maternal and nonmaternal rats were notedin other brain regions (e.g., ventromedial hypo-thalamic nucleus, BAOT, and anterior medial orposteroventral medial amygdaloid nuclei). Similarly,Fleming et al. (1994b) found that postpartum ratsexposed to pups showed many more FOS-IR neuronsin: MPOA, piriform cortex, and medial and corticalamygdala than mothers exposed to a familiar adultfemale or to food. The active display of maternalbehavior in lactating rats was also associated with anincrease in the number of fosB and egr-1 immuno-reactive neurons in theMPOAandventral bed nucleusof the stria terminalis, though the temporal course ofexpression of these proteins was different from thatobservedwith the c-FOS protein (Numan et al., 1998).The same group of investigators (Stack and Numan,2000) has shown that immunoreactivity to the c-FOSand fosB proteins is maintained in the MPOA, ventralbed nucleus of the stria terminalis, and PVN (anterior,magnocellular region) as long as mothers interact withtheir pups (maximal time studied was 47 h). In otherregions explored (e.g., posterodorsal medial amygdala,lateral habenula, posterior PVN, and ventromedialhypothalamic nucleus), immunoreactivity declinedover time despite continuous mother–litter interac-tion (Stack and Numan, 2000). Because the MPOAand ventral bed nucleus of the stria terminalis areessential for the display of maternal behavior in rats(see earlier), these authors have proposed that thecontinuous expression of the c-FOS and fosB genesmay be necessary to maintain the activity of theseareas. In addition, sensitized adult virgin rats alsoshowan increased number of neurons immunoreactiveto both c-FOS and fosB proteins in the MPOA andventral bed nucleus of the stria terminalis following a2-h interaction with pups (Kalinichev et al., 2000).

p0165Despite these clear results, the interpretation ofchanges in the number of FOS-immunoreactive neu-rons is not unequivocal because, in the context of abehavior that is stimulated by pup cues, a given brainregion may be activated by either: (1) the perceptionof such cues, (2) the expression of maternal behaviorper se, or (3) the occurrence of events not related withmaternal behavior (e.g., neuroendocrine secretion).To distinguish among these possibilities, strategies

10 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 11: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

have been used to selectively block the perception ofspecific cues from the pups and then assess theimpact of such manipulations on the expressionof FOS in particular brain regions. The removal ofnipples (thelectomy) did not modify the number ofFOS-IR neurons in the MPOA (Numan and Numan,1995; Walsh et al., 1996) or ventral bed nucleus of thestria terminalis (Numan and Numan, 1995) and theapplication of a local anesthetic in the ventrumreduced FOS expression only in the somatosensorycortex (Walsh et al., 1996). Similarly, the surgicalremoval of the olfactory bulbs (Numan and Numan,1995; Walsh et al., 1996) or the spraying of ZnSO4

into the olfactory epithelium (Fleming and Walsh,1994; Walsh et al., 1996) did not modify the numberof FOS-IR neurons in the MPOA of lactatingmothers but reduced it in structures related withthe olfactory system (e.g., medial amygdala and piri-form cortex). Moreover, the number of FOS-IR neu-rons in the MPOA following an interaction with thelitter remained unchanged even after the combinedapplication of local anesthetic to the ventrum plusZnSO4 to the olfactory epithelium (Fleming andWalsh, 1994; Walsh et al., 1996). However, the com-bined surgical removal of nipples and main olfactorybulbs reduced (but did not eliminate) the number ofFOS-IR neurons in the MPOA and ventral bednucleus of the stria terminalis (Numan and Numan,1995). From these results, it has been proposed thatneurons expressing FOS following an interactionwith the litter represent mostly the efferent outputfrom the MPOA and are related with the perfor-mance of maternal behavior (Numan and Numan,1995; Walsh et al., 1996). In a detailed study involvingthe quantification of FOS-immunoreactive neuronsin 20 brain regions, Lonstein et al. (1998) found thatthe degree of neuronal activation provoked by inter-acting with a suckling litter was not significantlydifferent from that observed following an interactionwith pups that had their snouts anesthetized andwere, therefore, unable to suckle. The single areawhere FOS expression was significantly greater fol-lowing suckling (vs. nonsuckling) stimulation wasthe caudal PAG (lateral and ventrolateral portions;Lonstein and Stern, 1997). Because only sucklingpups elicit the upright crouch (kyphosis) characteris-tic of normal nursing (Stern, 1996) and electrolyticlesions to the caudal PAG disrupt it (Lonstein andStern, 1997), these authors have suggested that adiscrete population of FOS-IR neurons in this regionis related with the sensorimotor control of kyphoticnursing. Indeed, unilateral thelectomy reduced both

the proportion of time spent in the kyphotic posture(in relation to the total time spent over the litter)and the number of FOS-IR neurons found in thecaudal PAG on the side ipsilateral to the thelectomy(Lonstein and Stern, 1999).

p0170To further understand the significance of changesin the number of FOS-IR neurons associated with thedisplay of maternal behavior, Lonstein and De Vries(2000) analyzed brain sections co-stained with FOSand an antiserum against GAD67 (an isoform of therate-limiting synthesizing enzyme for GABA, foundin the cytoplasm; Martin and Rimvall, 1993). Theyfound that, in lactating rats, interaction with pupsincreased the number of co-labeled neurons in theMPOA, ventral bed nucleus of the stria terminalis,and caudal PAG (ventrolateral region), a result sug-gesting that GABAergic neurons may promote spe-cific aspects of maternal behavior by removing tonicinhibitory influences. By using a similar experimen-tal strategy, Lonstein et al. (2000) reported that lac-tating rats allowed to interact with pups showed anincreased number of neurons co-stained with FOSand an antibody against the ERa than did mothersnot given young. These differences were particularlyevident in the MPOA, lateral habenula, and ventralbed nucleus of the stria terminalis.

p0175In virgin Balb/c mice a single brief (30min) expo-sure to pups increased the number of FOS-IR neuronsin the MPOA, corticomedial amygdala, entorhinaland piriform cortex, and anterior olfactory nucleus,regardless of whether females were intact or ovariec-tomized (Calamandrei and Keverne, 1994). Inter-estingly, the presentation of pups inside a dark,perforated box (that allowed females to perceiveonly olfactory and auditory signals from the young)stimulated FOS expression in the olfactory areas butnot in the MPOA. In contrast, the depletion of nor-adrenaline in the olfactory bulb (provoked by inject-ing 6-hydroxy-dopamine into the medial olfactorystria) reduced FOS expression only in the anteriorolfactory nucleus and the piriform cortex. Theseresults coincide with those obtained in rats and sup-port the notion that FOS expression in the MPOAoccurs as a result of the active display of maternalbehavior.

p0180In multiparous sheep, the interaction betweenthe mother and her lamb for the first 30min afterparturition stimulated c-fos mRNA transcription(detected by in situ hybridization) in the cerebralcortex (e.g., entorhinal, piriform, and somatosensory),habenula, hippocampus (CA3 region, dentate gyrus),limbic system (e.g., bed nucleus of the stria

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 11

HOBB: 00003

Page 12: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

terminalis, lateral septum, and olfactory bulb),MPOA, and several hypothalamic nuclei (e.g., PVNand SON). Very similar changes were observed innonpregnant multiparous ewes treated with estrogenand progesterone and given 5min of VCS. In contrast,zif-268 mRNA transcription was noted in only a fewareas (entorhinal and orbitofrontal cortex; dentategyrus) of postpartum sheep (Da Costa et al., 1997).

s0100 3.3.5.2 Other methodsp0185 The uptake of 14C-2-deoxy-glucose (2DG) has been

used as an index of activity in neuronal terminals butnot cell bodies (Nudo andMasterton, 1986; Sharp et al.,1988). At parturition, mother rats showed an increa-sed uptake of 2DG in the MPOA and VTA, while insensitized virgins displaying maternal behavior adecreased 2DG uptake was observed in several struc-tures of the accessory olfactory system (i.e., accessoryolfactory bulb, medial amygdaloid nucleus, and bednucleus of the accessory olfactory tract; Del Cerroet al., 1995). These results coincide with the proposi-tion that the facilitation of maternal behavior in virginrats occurs as a consequence of reducing the impact ofaversive olfactory stimulation coming from the pups.To distinguish between excitatory and disinhibitorysynaptic relations in specific brain areas, associatedwith the display of maternal behavior, the same groupof investigators combined the 2DG method with theimmunocytochemical detection of FOS (Komisaruket al., 2000). In the three models of maternal behaviorexplored (parturient, sensitized virgins, and rats hys-terectomized on pregnancyday 16), theMPOA showedincreased uptake of 2DG and increased expression ofFOS. This indicates that the expression of maternalbehavior (regardless of how it is induced) involvesexcitatory input to the MPOA and excitatory outputfrom it. A different pattern was observed in structuresof the olfactory system (e.g., BAOT and medial amyg-dala): while parturient rats showed increased 2DGuptake and increased FOS expression (i.e., excitatory/excitatory relations), in sensitized virgins 2DG uptakedecreased and FOS increased. This observation furthersupports the notion that the expression of maternalbehavior through sensitization in virgin rats involvesa reduction in the activity of the olfactory system.

p0190 To further explore the role of immediate earlygenes in the expression of maternal behavior, anti-sense oligodeoxyribonucleotides (ODNs) have beeninfused into the PVN of periparturient sheep (DaCosta et al., 1999). There is evidence that the releaseof OT from this nucleus at parturition promotesmaternal behavior in this species. The bilateral

infusion of antisense ODNs against c-FOS and c-jun

via microdialysis probes placed in the PVN antago-nized OT release in this region (but not in plasma) andreduced the expression of mRNA to specific peptides(e.g., OT, CRH, and pre-proenkephalin) also in thisnucleus. In addition, this procedure decreased the inci-dence of low-pitch bleats, though only at 10min post-partum. The other components of maternal behaviorwere not significantly modified and rejection beha-viors toward the neonate were not observed.

p0195In summary, the above studies detected discrete(if any) alterations in the maternal performance ofthe knockout mice used, supporting the notion thatthis complex behavior is regulated by a multitude ofhormones and neurotransmitter systems that act inconcert and may compensate for the lack of a partic-ular one. However, this does not rule out the possi-bility that a more primary element is essential for thecoordinated operation of such hormonal/neurotrans-mitter systems and, consequently, that its malfunctionor deletion may preclude the expression of mater-nal behavior. Indeed, work by Brown et al. (1996) pro-vided clear evidence to this effect. Homozygousfemale mice lacking the immediate early gene fosB

show a disruption of maternal behavior, failing toretrieve their pups and crouch over them, despitethe fact that they approach and sniff them. Surpris-ingly, such mothers show no alterations in spatiallearning, and their ability to discriminate betweentwo different odorants is similar to the one observedin wild-type animals. Moreover, pregnancy and par-turition are normal in such knockout mice and noalterations in the reproductive tract or mammaryglands are evident. Furthermore, the serum concen-trations of estradiol and progesterone during preg-nancy are also normal as are the levels of mRNA forOT in the hypothalamus and for PRL in the pituitary.Interestingly, the deleterious effects of the fosB muta-tion on maternal behavior are not overcome by expe-rience: the lack of maternal responsiveness is stillobserved after successive pregnancies and parturi-tions. Additionally, virgin fosB knockouts (males andfemales) are also unable to respond maternally tofoster pups, even after repeated exposure to them.

s01053.4 Epigenetic, IntergenerationalTransmission of Mothering Styles:Its Impact on Offspring Development

p0200Over the last 10 years a large body of research hasinvestigated the intriguing observation that the

12 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 13: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

environment surrounding a developing organism canpermanently affect its performance in adulthood.Although this issue has been well recognized sincethe early studies of Levine (1957) and Denenberg(1964), recent studies have revealed the multiplelevels at which the organism’s phenotype is modifiedby early experience (e.g., neuroendocrine reactivity,cognition, social behavior, and sensory gating), theways by which the environment can act on thegenome to modify the brain of the developing indi-vidual (epigenetic), and how the characteristicsacquired in infancy can be transmitted to the nextgeneration (intergenerational). At the core of thisresearch lies the assumption that, even in those spe-cies in which the young are born precocial, the fine-tuning of the circuits mediating complex tasks isunfinished and, therefore, subject to permanent mod-ification by environmental influences. The existenceof critical periods for a variety of traits has beenacknowledged for many years ( Johnson, 2005; Micheland Tyler, 2005), for example, in studies exploringthe role of steroids and tactile stimulation in orient-ing brain sexual differentiation (Beyer and Feder,1987; McCarthy and Konkle, 2005; Moore, 1995) orthe learning by young animals of odors and flavorsthat will determine their mating and food preferencesin adulthood (reviewed in Fleming et al. (2002)).These studies have revealed the existence of timewindows during which the exposure or not of adeveloping individual to specific forms of stimulation(e.g., tactile, olfactory, and hormonal) will determinethe direction and magnitude of a given response inadulthood.

p0205 By contrast, little is known about the mechanismsthat mediate the translation between the perceptionof such varied forms of stimulation and the perma-nent modification of brain function. In the last 7 years,work coming mainly from the laboratory of MichaelMeaney has provided evidence for the operation ofspecific pathways between the tactile stimulationprovided by mother rats to their litter and the adultphenotype of their offspring (see below). The experi-mental strategy used in these studies has been theselection of mother rats according to the frequencywith which they lick the pups’ body. This behavior isa normal component of rat maternal care, and itsmagnitude within the population follows a normaldistribution (Champagne et al., 2003). By choosingthe extremes of the population (i.e., mothers locatedone standard deviation apart from the mean in bothdirections), Meaney’s group has found that mothersproviding high levels of licking (and arched-back

nursing) during the first week of life breed offspringthat, in adulthood, show reduced reactivity of thehypothalamic–pituitary–adrenal (HPA) axis to stress-ful situations, decreased fear responses, a greatercapacity for spatial learning, and a high frequency oflicking toward their own offspring. These behavioralcharacteristics are associated with a higher concen-tration of glucocorticoid receptors (GRs) in the hip-pocampus, decreased hypothalamic CRF expression,larger concentrations of ERa in the MPOA, largernumbers of OT receptors in specific diencephalicregions, and more GABAA-benzodiazepine receptorsin several amygdaloid nuclei than the offspring ofmothers that lick their pups less frequently. Suchdifferences are not due to the genetic characteristicsof the mothers’ offspring as cross-fostering studieshave shown that the adult phenotype correspondsto that of the rearing mother, rather than to thebiological one. Moreover, by studying the maternalbehavior shown as adults by the female offspring ofthe two types of selected rats, the mothering style wasthat of the rearing mother. In other words, an inter-generational transmission of acquired behavioraltraits had occurred.

p0210The way in which alterations in specific receptorsmay relate to differences in behavior has beeninvestigated in relation to the stress response. Thehippocampus of female offspring from high licking/arched-back nursing mothers shows a higher expres-sion of cyclic AMP-response element binding protein(CREB)-binding protein (CBP), a higher acetylationof histones, and a reduced DNA methylation patternof the GR exon 17 promoter in the hippocampus (forcomprehensive reviews, see Champagne et al. (2003),Szyf et al. (2005), and Weaver et al. (2004)). Takentogether, the findings from Meaney’s group haverevealed a specific pathway by which maternal stim-ulation, received at a critical period, can permanentlymodify the expression of particular receptors (GRs)in the brain which, in turn, play a crucial role inthe way adult animals respond to stress. Whethersimilar mechanisms operate in relation to other beha-viors, receptors, or neurotransmitters remains to beinvestigated.

p0215Different experimental strategies have been usedby other investigators to assess the impact of theneonatal environment in determining the adult char-acteristics of the offspring: (1) short (c. 15min day–1;the so-called handling paradigm) versus long mother–young separations; (2) total maternal deprivationaccompanied by artificial rearing; (3) natural altera-tions of maternal behavior provoked by stressful

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 13

HOBB: 00003

Page 14: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

surroundings (e.g., as a consequence of social inter-actions); and (4) alterations of the nest environment(e.g., physical materials, and siblings). These studieshave taken into account the biology of the species,that is, the pattern of maternal care, the number anddevelopmental state of the offspring born. The firsttwo strategies have been tried in several strains of ratsand, in general, the handling procedure has beenfound to improve some characteristics of the off-spring when compared with control groups notremoved from their mothers. Thus, as adults, hand-led animals show: reduced emotionality, enhancedlearning, and better maternal responsiveness in adult-hood (Rees and Fleming, 2001). These effects are aconsequence of changes in the mother’s behaviorupon reunion with the litter, such as increased lickingand grooming of the pups, which, in turn, result fromthe increased demands (ultrasonic vocalizations,suckling, and rooting) made by the young (Pereiraand Ferreira, 2006).

p0220 By contrast, long mother–young separations(3–5 h day–1 for at least the first 7 postnatal days)provoke the reverse, that is, increased fear and emo-tional responses, deficient maternal care in adult-hood, and impairments in specific forms of learning(Fleming et al., 1999, 2002; Lovic et al., 2001). Theseresults agree with the findings from Meaney’s groupand emphasize: (1) the importance of specific formsof maternal tactile stimulation for the programmingof certain adult characteristics and (2) the existenceof critical periods during which the plasticity of thecentral nervous system (CNS) is particularly sensi-tive to environmental influences. Indeed, by usingthe maternal separation/artificial rearing strategy,Fleming and colleagues found severe alterations inemotionality (exaggerated responses to stress), sen-sory gating (revealed by a deficiency in prepulsestartle inhibition), and social recognition (evidencedthrough deficits in olfactory memory), in addition tothe expression of behaviors indicative of anxiety(Fleming et al., 1999, 2002; Lovic et al., 2001). Thefinding that intense anogenital stimulation providedwith a brush, exposure to nest odor, or rearing with alittermate can partially or totally reverse some ofthose shortcomings reveals that the plasticity of theCNS during infancy can follow several directions(Melo et al., 2006).

p0225 Altering the immediate environment to the extentthat it creates stress in pregnant or lactating mothersis sometimes the outcome of housing in farm animals(Moberg and Mench, 2001) or of the establishment ofsocial hierarchies in some species (notably, primates).

Sows, ewes, and rhesus macaques have been studiedunder these conditions, in terms of their own mater-nal behavior while under stress and in relation to theconsequences for their offspring. Detrimental effectsof stress during gestation were manifest in sows asincreased cortisol secretion and smaller increasesin body weight across pregnancy. The daughters ofthese sows, when tested as adults, showed abnormalmaternal behavior toward their own piglets, increa-sed cortisol secretion in response to social stress, andan enhanced expression of CRH mRNA in the PVN( Jarvis et al., 2006). Lambs prevented from sucklingtheir mothers (and receiving milk from a bucket) showlarger cortisol secretion, more distress bleats, less timenear their companions, and reduced growth, com-pared with dam-suckled lambs (Napolitano et al.,2003). Deficient contact between ewe and lambin early lactation may have consequences; it is un-known, however, if such early developmental effectsmodify the adult maternal behavior of those femaleoffspring.

p0230The maternal behavior of rhesus macaques kept incolonies under seminatural conditions varies fromabusive to protective; these characteristics are main-tained within individuals across successive parturi-tions. Abusive mothers are characterized by highrates of rejections and contact-breaking from theirinfants. Maestripieri and colleagues found that thispattern of maternal care has a profound impact onoffspring development. Abused infants show higherrates of distress vocalizations and anxiety, later inde-pendence from their mothers, and abnormal playbehavior, compared with infants from nonabusivemothers (McCormack et al., 2006). Moreover, cross-fostered females, tested as adults, resembled theirfoster mothers in the rates of maternal rejectiontoward their own infants but were more like theirbiological mothers in their contact-making behavior(Maestripieri et al., 2007). Abusive mothers also hadlower levels of a serotonin metabolite (5-hydroxy-indole-acetic acid – 5-HIAA) in the cerebrospinalfluid than did the nonabusive ones. In a differentexperimental model the behavior of rhesus monkeysreared with same-aged peers was compared to that ofmonkeys reared by mothers. As adults, peer-rearedfemales were more likely to reject or abuse theirinfants, were more reactive to stress, more aggressivein social interactions, and exhibited lower basal cate-cholamine system activity (for review, see Fleminget al. (1999)).

p0235Unlike most rodents guinea pigs are born preco-cial, that is, covered with body hair, able to regulate

14 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 15: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

their body temperature, with their eyes open, andable to urinate and defecate without anogenital stim-ulation from their mother. Despite these character-istics, young guinea pigs depend on their mother forthe regulation of HPA axis reactivity. When placed ina new environment, the mere presence of the motherbuffers this stressful condition; cortisol release is lessand distress vocalizations are fewer (Hennessy, 2003;Wewers et al., 2003) than if the mother is absent. Theimpact of repeated mother–young separation on thebehavior, emotional reactivity, and maternal behaviorof the offspring is unknown.

p0240 Rabbit pups, despite being born altricial, havelittle contact with their mother from parturition andthroughout lactation except for the 3-min dailynursing bout (see Section 3.2). Yet, they remain inclose contact with each other inside the maternalnest, exposed to its rich olfactory environment, andprotected from harsh temperatures and predators.The relevance of this immediate environment,which hardly involves the mother, for the develop-ment of rabbit pups was investigated by isolating theyoung from each other and bringing them togetheronly for nursing. This procedure compromised thedevelopment of thermoregulation, when comparedwith pups that were kept together in a huddle(Bautista et al., 2003).

p0245 The information summarized in this section showsthat in all mammals studied the maternal careprovided to the offspring has a profound impact onthe development of many complex functions, includ-ing emotional reactivity, social interactions, foodselection, spatial learning, mate choice, thermoregu-lation, etc. Moreover, specific studies have revealedthat certain characteristics of maternal behavior itselfare permanently altered in the female offspring, thusoffering a means for the intergenerational transmis-sion of such acquired traits. Yet, when comparing theresults of particular studies that explored the sameissue, discrepancies abound. For instance, the impactof long mother–young separations on maternalbehavior of the adult female offspring varies depend-ing on whether: they were tested as sensitized virginsor lactating mothers; they were exposed to young asjuveniles or not; and the pups given were their own oralien ones (Darnaudery et al., 2004). The test selectedto measure reactivity to a new environment (e.g.,behavior in the elevated plus maze (EPM) vs. cortisolsecretion vs. ambulation in an open field), the sexof the individual studied (more studies have beenperformed in females than in males; Barna et al.,2003), and, of course, the biology of the species

under study all determine the outcome of a particularstudy. Clearly, much more work is needed in thisexciting new field to reveal the extent of maternaleffects on offspring development and the multiplicityof mechanisms that may participate in the trans-mission of specific behavioral and neuroendocrinecharacteristics across generations via an epigeneticroute.

s01103.5 Impact of Maternal Behavior onthe Mother: Effects on Cognition andNeuroplasticity

s01153.5.1 Theoretical Context

p0250The new mother is a study in contrasts. She is thesame female she was before the birth of her offspring;conversely, the transition from nulliparous to parousfemale involves substantial, likely permanent, altera-tions to both brain and behavior. In this section wedetail some of these changes and their ramifications.These questions arise naturally from the simple rec-ognition of the requisite behavioral set for thenew mother, including both behaviors directlyrelated to pup care and maintenance (the so-calledmaternal behavior), and those that are ancillary toor supportive of maternal behavior and require themother to be efficient in her search for resources(Kinsley et al., 1999; Lambert et al., 2005). We discussthe focus of these changes and activities, the off-spring, who represent the culmination of a lifetimeof metabolic investment and the mother’s geneticlegacy.

p0255The mother is confronted with a stark lose–losesituation. She can stay on her nest, safe with heroffspring, and face eventual starvation and the lossof her litter. Or she can leave the nest and her vul-nerable brood in search of food and water, in whichcase she places both herself and those young in dan-ger. We predicted that the mother would benefit frompositive alterations to behaviors that underlie theaforementioned activities. That is, departing the safeconfines of the nest for environments and dangersunknown requires overcoming the fear inherent todoing so, and successfully confronting any risksencountered (predators, rogue males, impassableobstacles, etc.). Then, any foraging costs (increasedtime and effort) could be reduced by enhancementsto spatial and reference memory. These latter chan-ges would ensure a rapid return to the nest andthe defenseless offspring. Therefore, reductionsin fear and anxiety, and improvements to spatial

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 15

HOBB: 00003

Page 16: HOBB: 00003 - Elsevier
Page 17: HOBB: 00003 - Elsevier
Page 18: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

pathways and their downstream alteration of neuro-nal structure and function, as suggested by Macbethet al. (2008). Such offspring-enriching effectswere suggested by Kinsley et al. (1999) and reportedby Lambert et al. (2005) and identify an interestingform of symbiosis in which the offspring have ahand/paw in their own survival and bettermentthrough stimulation of their mother’s brain, and herbehavior toward them. Parrish et al. (2007) providean interesting model by which dendritic connec-tions and networks could be created/maintained bysuch rich translation of the environment into neuralcomplexity.

p0295 Reproductive experience does modify the female’sbrain; further, that the brain owes some of its plas-ticity to the hormonal environment has been welldocumented. The MPOA, whose circuitry and in-volvement in maternal behavior has been describedvoluminously, exhibits marked c-fos expression fol-lowing pup interactions and treatments (opiates) thatmodify normal maternal motivation (Numan andInsel, 2003; Stafisso-Sandoz et al., 1998). ObservingGolgi-stained neurons, Keyser-Marcus et al. (2001)have reported that perikarya volume and neuronalcomplexity increased in the MPOA of late-pregnant(day 21), lactating (day 5), and pregnancy hormone-treated (sequential estradiol and progesteroneimplants) females. This increased activity associatedwith the onset of maternity suggests a region of thebrain that is preparing or is ready to display therequisite behaviors toward the pups. The increase insize/volume of the cell body, where a majority ofprotein syntheses are occuring (Miller and Erskine,1995), provides a neurophysiological concomitant,both metaphorically and literally, to the upregulationof activity necessary for successful reproduction.

p0300 In addition to OT, steroid hormones modify boththe structure and function of the hippocampus.Short-term cyclic fluctuations in steroid hormonessuch as estradiol and progesterone can modulate theconcentrations of CA1 hippocampal dendritic spines(Gould et al., 1990; Li et al., 2004; Woolley, 1998;Woolley et al., 1990, 1996; Woolley and McEwen,1992, 1993; Yankova et al., 2001). This researchfocused on nulliparous female rats (although somemales (in this case primates) may show interestingparallels (Kozorovitskiy et al., 2006)).

p0305 As alluded to above, there are likely two primaryroutes by which the altered behavioral capacities ofthe parous female occur, the hormones of pregnancyand the significant stimulation provided by the pups.For example, female rats undergo cyclical and

sizeable hormonal (primarily estradiol and progester-one) alterations during the 4–5-day estrous cycle. Onthe other hand, pregnancy involves substantially ele-vated estrogens and progestins for a significantlylonger duration and with considerably elevated levelscompared to the estrous cycle (e.g., days vs. hours,and higher absolute estrogen levels; during mid–latepregnancy (days 10–22), these levels more than dou-ble their earlier (days 1–10) levels, reaching a peakof approximately 80 pg ml–1 on days 18–22; Bridges,1984).

p0310Enhancing these potent steroid hormones and theiranabolic properties predicts strong and enduringdownstream effects. Therefore, given the markedincreases in these steroid hormones, one might expecteven greater effects on CA1 dendritic spine prolifera-tion as the female experiences the hormonal cocktailthat is pregnancy. Kinsley et al. (2006) found thatspine density was increased in late-pregnant andlactating (day 5) females (which were not differentfrom each other) compared to the other virgingroups, including proestrous females, who had morespines than diestrous and estrous animals (which hasbeen documented previously), and OVX femalestreated with a pregnancy-mimicking (and maternalbehavior-stimulating; Bridges, 1984) sequential estra-diol plus progesteorne hormonal regimen. Such ani-mals had significantly more dendritic spines thanOVX control females (and had numbers that werenot significantly from those of the late-pregnant andlactating females). Pawluski and Galea (2005)reported some lingering effects of parity on hippo-campal neuronal morphology, with the primiparousfemales having significantly greater dendritic arborsafter weaning their litters. The hippocampus, there-fore, appears sensitive and responsive to the hor-mones of pregnancy. The major question is Why?,and it appears to be answered by the behavioraleffects noted above and in Macbeth et al. (2008) inwhich foraging, spatial, and other forms of memoryare enhanced, supporting the display of maternalbehavior. Rasia-Filho et al. (2004) have reported sim-ilar neuronal structural effects in the amygdala ofparous females, suggesting differential neuronalinteractions therein, and consequent alterations ofregulation of fear and emotion – behaviors thatare reliably reported as reduced in parous females(Wartella et al., 2003).

p0315Although the hippocampus may not play a directrole in care of the young (viz., full pup-directed mater-nal behavior; but see also Numan and Insel (2003)),it may maintain ancillary maternal behaviors and

18 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 19: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

responses to the environment, other examples ofneuroplasticity that collectively ensure successfulreproduction. For example, there is evidence of estra-diol-induced hippocampal flexibility across species,including increases in synapse-associated proteinimmunoreactivity (Choi et al., 2003; Li et al., 2004);enhancements in long-term potentiation and learning(Warren et al., 1995; Warren and Juraska, 1997); up-regulation of NMDA receptors (Adams et al., 2004);downregulation of reactive astroglia (Garcia-Estradaet al., 1993, 1999); increases in synaptophysin (Fricket al., 2002); and decreases in experimentally inducedapoptosis (Liu et al., 2001). Another interesting fea-ture of hippocampal plasticity is the association ofestrogen status with BDNF mRNA and immuno-reactivity (Solum and Handa, 2002; Scharfman et al.,2003) – data that connect to the Macbeth et al. (2008)observations reported above.

p0320 Recent work from our lab, using reverse transcrip-tase polymerase chain reaction (RT-PCR) (Continoet al., 2007), has implicated a number of hippocampaland prefrontal cortical (PFC) neuroplastic genes inthe expression of the maternal brain. For example, anupregulation of the genes for BDNF, its receptorNTrk2, syntaxin (a presynaptic protein), spinophilin(a dendritic spine regulator), Cox-1 (a mitochondrialprotein); and amyloid precursor protein in the PFCall show interesting differences between parous andnulliparous females. When taken in total, it seemsreasonable to posit that reproduction represents asignificant event in the life of the female. Thechanges are substantial and enduring, and are on apar with other developmental milestones such assexual differentiation and puberty. The data suggestthat the hormones of pregnancy interact with post-partum pup exposure to produce an enriched envi-ronment and thereby improve learning ability (atleast temporarily and possibly permanently), mitigateanxiety and stress responsiveness, and enhance prob-lem solving in a novel context (integration of cogni-tive and stress responses), in the mother. It can beargued that such effects would be selected for, andwould fulfill the criterion of incrementally adaptivechanges favored by natural selection (Kinsley andLambert, 2006).

p0325 Finally, it may be interesting to look at thechanges to a single and singular sense, olfaction, andits potential role in the transition from nulliparous toparous female. Macbeth et al. (2008) made tissuepunches from the olfactory bulb and examinedneurotransmitter dynamics therein. We know thatthe rat is such an olfactory-guided animal, and that

its maternal behavior is substantially regulated byolfactory cues from young (see Fleming andRosenblatt (1974a,b)). Macbeth et al. (2008) reportedincreased concentrations in the olfactory bulb ofdopamine (DA) and its metabolite 3,4-dihydroxy-phenylacetic acid (DOPAC); norepinephrine (NE)and its metabolite 3-methoxy-4-hydroxyphenylglycol(MHPG); and the serotonin (5-HT) metabolite5-HIAA, in multiparous as compared to nulliparousfemales. These data indicate the possibility of altera-tions to upfront olfactory regulation of behavior,with resulting modifications of behavioral responses.Furthermore, turnover ratios, as defined by Macbethet al. (2008) as metabolite-to-monoamine ratios, alsodemonstrated some interesting parity effects, withmultiparous females having significantly higher turn-over ratios of DOPAC:DA and 5-HIAA:5HT com-pared to nulliparous females, but a lower turnoverratioth

Page 20: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

the tonic inhibition exerted by the olfactory systemin species other than rodents); more hormones havebeen added to the cohort that stimulates the onsetand/or maintenance of maternal responsiveness (i.e.,corticosteroids); and our knowledge about the neuro-endocrine control of maternal behavior in farm ani-mals has increased. Still, many key issues surroundingthe complex behavioral and neuroendocrine transi-tion across estrus, pregnancy, parturition, and lacta-tion remain uninvestigated. For instance, althoughall rodents display nest-building, the hormonal con-trol of this activity and the brain regions involvedin its expression are largely unknown. Similarly, thequantitative and qualitative changes in food intakeobserved in many mammals around parturition(including placentophagia) have been little explored,either in terms of their potential biological meaningor the factors that control them. Indeed, the peri-partum stage is increasingly being recognized as acritical period for both mother and young, during

Page 21: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Algers B and Uvnas-Moberg K (2007) Maternal behavior in pigs.Hormones and Behavior 52: 78–85.

Andrews ZB and Grattan DR (2002) Opiod control of prolactinsecretion in late pregnant rats is mediated bytuberoinfundibular dopamine neurons. Neuroscience Letters328: 60–64.

Avar Z and Monos E (1966) Effect of lateral hypothalamic lesionon pregnant rats and foetal mortality. Acta medicaAcademiae Scientiarum Hungaricae 22: 259–264.

Avar Z and Monos E (1967) Effect of lateral hypothalamiclesion on maternal behavior and foetal vitality in the rat.Acta medica Academiae Scientiarum Hungaricae 23:255–261.

Avar Z and Monos E (1969a) Behavioural changes in pregnantrats following far-lateral hypothalamic lesions. Acta medicaAcademiae Scientiarum Hungaricae 35: 295–303.

Avar Z and Monos E (1969b) Biological role of lateralhypothalamic structures participating in the control ofmaternal behavior in the rat. Acta medica AcademiaeScientiarum Hungaricae 35: 285–294.

Bakowska JC and Morrell JI (1997) Atlas of neurons thatexpress mRNA for the long form of the prolactin receptor inthe forebrain of the female rat. Journal of ComparativeNeurology 386: 161–177.

Barna I, Balint E, Baranyi J, Bakos N, Makara GB, and Haller J(2003) Gender-specific effect of maternal deprivation onanxiety and corticotrophin-releasing hormone mRNAexpression in rats. Brain Research Bulletin 62: 85–91.

Barone FC, Weiner MJ, Scharoun SL, Guevara-Aguilar R, andAguilar-Baturoni HU (1981) Afferent connection of the lateralhypothalamus: A horseradish peroxidase study in the rat.Brain Research Bulletin 7: 75–88.

Bautista A, Drummond H, Martınez-Gomez M, and Hudson R(2003) Thermal benefit of sibling presence in the newbornrabbit. Developmental Psychobiology 43: 208–215.

Beach FA (1937) The neural basis of innate behavior. I. Effectsof cortical lesions upon the maternal behavior pattern in therat. Journal of Comparative and Physiological Psychology 24:393–436.

Beach FA and Jaynes J (1956) Studies of maternal retrieving inrats. III. Sensory cues involved in the lactating female’sresponse to her young. Behaviour 10: 104–125.

Benuck I and Rowe FA (1975) Centrally and peripherallyinduced anosmia: Influences on maternal behavior inlactating female rats. Physiology and Behavior 14: 439–447.

Beyer C and Feder HH (1987) Sex steroids and afferent input:Their roles in brain sexual differentiation. Annual Review ofPhysiology 49: 349–364.

Blaustein JD, King JC, Toft DO, and Turcotte J (1988)Immunocytochemical localization of estrogen-inducedprogestin receptors in guinea pig brain. Brain Research474: 1–15.

Blaustein JD and Turcotte JC (1989) Estradiol-inducedprogestin receptor immunoreactivity is found only inestrogen receptor-inmunoreactive cells in guinea pig brain.Neuroendocrinology 49: 454–461.

Bodensteiner KJ, Cain P, Ray AS, and Hamula LA (2006) Effectsof pregnancy on spatial cognition in female hoodedLong-Evans rats. Hormones and Behavior 49: 303–314.

Bridges RS (1975) Long-term effects of pregnancy andparturition upon maternal responsiveness in the rat.Physiology and Behavior 14: 245–249.

Bridges RS (1977) Parturition: Its role in the long termretention of maternal behavior in the rat. Physiology andBehavior 18: 487–490.

Bridges RS (1984) A quantitative analysis of the roles of dosagesequence and duration of estradiol and progesteroneexposure in the regulation of maternal behavior in the rat.Endocrinology 114: 930–940.

Bridges RS and Freemark MS (1995) Human placentallactogen infusions into the medial preoptic area stimulatematernal behavior in steroid-primed, nulliparous female rats.Hormones and Behavior 29: 216–226.

Bridges RS and Grimm CT (1982) Reversal of morphinedisruption of maternal behavior by concurrenttreatment with the opiate antagonist naloxone. Science218: 166–168.

Bridges RS and Mann PE (1994) Prolactin–brain interactionsin the induction of material behavior in rats.Psychoneuroendocrinology 19: 611–622.

Bridges RS, Numan M, Ronsheim PM, Mann PE, and Lupini CE(1990) Central prolactin infusions stimulate maternalbehavior in steroid-treated, nulliparous female rats.Proceedings of the National Academy of Sciences of theUnited States of America 87: 8003–8007.

Bridges RS, Robertson MC, Shiu RP, Sturgis JD,Henriquez BM, and Mann PE (1997) Central lactogenicregulation of maternal behavior in rats: Steroid dependence,hormone specificity, and behavioral potencies of ratprolactin and rat placental lactogen I. Endocrinology 138:756–763.

Broad KD, Kendrick KM, Sirinathsinghji DJ, and Keverne EB(1993) Changes in oxytocin immunoreactivity and mRNAexpression in the sheep brain during pregnancy, parturitionand lactation and in response to oestrogen andprogesterone. Journal of Neuroendocrinology 5: 435–444.

Broad KD, Levy F, Evans G, Kimura T, Keverne EB, andKendrick KM (1999) Previous maternal experiencepotentiates the effect of parturition on oxytocin receptormRNA expression in the paraventricular nucleus. EuropeanJournal of Neuroscience 11: 3725–3737.

Brown JR, Ye H, Bronson RT, Dikkes P, and Greenberg ME(1996) A defect in nurturing in mice lacking the immediateearly gene fosB. Cell 86: 297–309.

Bucher KL (1970) Temporal Lobe Neocortex and MaternalBehavior in Rhesus Monkeys. PhD Thesis, School of Hygieneand Public Health. Baltimore, MD: Johns Hopkins University.

Buckwalter JG, Stanczyk FZ, McCleary CA, et al. (1999)Pregnancy, the postpartum, and steroid hormones: Effectson cognition and mood. Psychoneuroendocrinology 24:69–84.

Burne THJ (2000) Expression of PGF2-alpha receptor mRNAin the female pig brain. European Journal of Neuroscience12(supplement 11): 407.

Caba M, Beyer C, Gonzalez-Mariscal G, and Morrell JI (2003a)Immunocytochemical detection of estrogen receptor alphain the female rabbit forebrain: Topography and regulation byestradiol. Neuroendocrinology 77: 208–222.

Caba M, Rovirosa MJ, Beyer C, and Gonzalez-Mariscal G(2003b) Immunocytochemical detection of progesteronereceptor in the female rabbit forebrain: Distribution andregulation by oestradiol and progesterone. Journal ofNeuroendocrinology 15: 855–864.

Calamandrei G and Keverne EB (1994) Differential expression ofFos protein in the brain of female mice dependent on pupsensory cues and maternal experience. BehavioralNeuroscience 108: 113–120.

Carlson NR and Thomas GJ (1968) Maternal behavior of micewith limbic lesions. Journal of Comparative and PhysiologicalPsychology 66: 731–737.

Champagne FA, Francis DD, Mar A, and Meaney MJ (2003)Variations in maternal care in the rat as a mediating influencefor the effects of environment on development. Physiologyand Behavior 79: 359–371.

Chirino R, Beyer C, and Gonzalez-Mariscal G (2007) Lesionto the main olfactory epithelium facilitates maternalbehavior in virgin rabbits. Behavioural Brain Research 180:127–132.

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 21

HOBB: 00003

Page 22: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Choi JM, Romeo RD, Brake WG, Bethea CL, Rosenwaks Z, andMcEwen BS (2003) Estradiol increases pre- and post-synaptic proteins in the CA1 region of the hippocampus infemale rhesus macaques (Macaca mulatta). Endocrinology144: 4734–4738.

Conrad LCA and Pfaff DW (1976) Efferences from medial basalforebrain and hypothalamus in the rat. I. Medial preopticarea. Journal of Comparative Neurology 169: 185–220.

Consiglio AR and Lucion AB (1996) Lesion of hypothalamicparaventricular nucleus and maternal aggressive behavior infemale rats. Physiology and Behavior 59: 591–596.

Contino R, Friedenberg J, Christon L, et al. (2007) Theexpression of the maternal brain: Specific genes involved inthe neuroplasticity of motherhood. Society for Neuroscience,Abstract 309: 5.

Corodimas KP, Rosenblatt JS, Canfield ME, and Morrell JI(1993) Neurons in the lateral subdivision of the habenularcomplex mediate the hormonal onset of maternal behavior inrats. Behavioral Neuroscience 107: 827–843.

Cruz ML and Beyer C (1972) Effects of septal lesions onmaternal behavior and lactation in the rabbit. Physiology andBehavior 9: 361–365.

Dacoit JP, Squinto SP, and Bazan NG (1990) Fos-jun and theprimary genomic response in the nervous system.MolecularNeurobiology 2: 27–55.

Da Costa AP, Broad KD, and Kendrick KM (1997) Olfactorymemory and maternal behaviour-induced changes in c-fosand zif/268 mRNA expression in the sheep brain. BrainResearch – Molecular Brain Research 46: 63–76.

Da Costa AP, De La Riva C, Guevara-Guzman R, andKendrick KM (1999) C-fos and c-jun in the paraventricularnucleus play a role in regulating peptide gene expression,oxytocin and glutamate release, and maternal behaviour.European Journal of Neuroscience 11: 2199–2210.

Da Costa AP, Guevara-Guzman RG, Ohkura S, Goode JA, andKendrick KM (1996a) The role of oxytocin release in theparaventricular nucleus in the control of maternal behaviourin the sheep. Journal of Neuroendocrinology 8: 163–177.

Da Costa AP, Wood S, Ingram CD, and Lightman SL (1996b)Region-specific reduction in stress-induced c-fos mRNAexpression during pregnancy and lactation. Brain Research742: 177–184.

Darnaudery M, Koehl M, Barbazanges A, Cabib S, LeMoal M,and Maccari S (2004) Early and later adoptionsdifferently modify mother–pup interactions. BehavioralNeuroscience 118: 590–596.

Del Cerro MC, Izquierdo MA, Collado P, Segovia S, andGuillamon A (1991) Bilateral lesions of the bed nucleus of theaccessory olfactory tract facilitate maternal behavior in virginfemale rats. Physiology and Behavior 50: 67–71.

Del Cerro MC, Izquierdo MA, Rosenblatt JS, Johnson BM,Pacheco P, and Komisaruk BR (1995) Brain 2-deoxyglucoselevels related to maternal behavior-inducing stimuli in the rat.Brain Research 696: 213–220.

Denenberg VH (1964) Critical periods, stimulus input, andemotional reactivity: A theory of infantile stimulation.Physiological Reviews 71: 335–351.

Denenberg VH, Taylor RE, and Zarrow MX (1969) Maternalbehaviour in the rat: An investigation and quantification ofnest building. Behaviour 34: 1–16.

Di Carlo R and Muccioli G (1981) Presence of specificprolactin binding sites in the rabbit hypothalamus. LifeSciences 28: 2299–2307.

Douglas AJ, Johnson H, Brunton P, and Russell JA (2000) BrainOT: Differential inhibition of neuroendocrine stressresponses and anxiety-related behaviour in virgin, pregnantand lactating rats. Journal of Neuroendocrinology 12:343–350.

Douglas AJ, Johnstone HA, Wigger A, Landgraf R, Russell JA,and Neumann ID (1998) The role of endogenous opioids inneurohypophysial and hypothalamo-pituitary–adrenal axishormone secretory responses to stress in pregnant rats.Journal of Endocrinology 158: 285–293.

Erskine MS, Barfield RJ, and Goldman BD (1978) Intraspecificfighting during late pregnancy and lactation in rats andeffects of litter removal. Behavioral Biology 23: 206–218.

Fahrbach SE, Morrell JI, and Pfaff DW (1985) Possible role forendogenous OT in estrogen-facilitated maternal behavior inrats. Neuroendocrinology 40: 526–532.

Fahrbach SE and Pfaff DW (1986) Effect of preoptic regionimplants of dilute estradiol on the maternal behavior ofovariectomized, nulliparous rats. Hormones and Behavior20: 354–363.

Felicio LF and Canteras N (2008) Maternal choices: Neuralmediation – caring for young or hunting?. In: Bridges RS (ed.)Neurobiology of the Parental Brain, pp. 75–82. San Diego,CA: Academic Press.

Fleischer S and Slotnick BM (1978) Disruption of maternalbehavior in rats with lesions of the septal area. Physiologyand Behavior 21: 189–200.

Fleming AS, Kraemer GW, Gonzalez A, Lovic V, Rees S, andMelo A (2002) Mothering begets mothering: Thetransmission of behavior and its neurobiologyacross generations. Pharmacology, Biochemistry, andBehavior 73: 61–75.

Fleming AS, Miceli M, and Moretto D (1983) Lesions of themedial preoptic area prevent the facilitation of maternalbehavior produced by amygdala lesions. Physiology andBehavior 31: 503–510.

Fleming A, Morgan HD, and Walsh C (1996) Experiential factorsin postpartum regulation of maternal care. In: Rosenblatt JSand Snowdon CT (eds.) Advances in the Study of Behavior,vol. 25, pp. 295–332. San Diego, CA: Academic Press.

Fleming AS, O’Day DH, and Kraemer GW (1999) Neurobiologyof mother–infant interactions: Experience and centralnervous system plasticity across developmentand generations. Neuroscience and Biobehavioral Reviews23: 673–685.

Fleming AS and Rosenblatt JS (1974a) Olfactory regulation ofmaternal behavior in rats. I. Effects of olfactory bulb removalin experienced and inexperienced lactating and cyclingfemales. Journal of Comparative and PhysiologicalPsychology 86: 221–232.

Fleming AS and Rosenblatt JS (1974b) Olfactory regulation ofmaternal behavior in rats. I. Effects of peripherally inducedanosmia and lesions of the lateral olfactory tract inpup-induced virgins. Journal of Comparative andPhysiological Psychology 86: 233–246.

Fleming AS, Suh EJ, Korsmit M, and Rusak B (1994) Activationof Fos-like immunoreactivity in the medial preoptic areaand limbic structures by maternal and social interactions inrats. Behavioral Neuroscience 108: 724–734.

Fleming AS, Vaccarino F, and Luebke C (1980) Amygdaloidinhibition of maternal behavior in the nulliparous female rat.Physiology and Behavior 25: 731–743.

Fleming AS and Walsh C (1994) Neuropsychology of maternalbehavior in the rat: c-fos Expression during mother–litterinteractions. Psychoneuroendocrinology 19: 429–443.

Franz JR, Leo RJ, Steuer MA, and Kristal MB (1986) Effects ofhypothalamic knife cuts and experience on maternalbehavior in the rat. Physiology and Behavior 38: 629–640.

Franzen EA and Myers RE (1973) Neural control of socialbehavior: Prefrontal and anterior temporal cortex.Neuropsychologia 11: 141–157.

Frick KM, Fernandez SM, and Bulinski SC (2002) Estrogenreplacement improves spatial reference memory and

22 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 23: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

increases hippocampal synaptophysin in aged female mice.Neuroscience 115: 547–558.

Gaffori O and Le Moal M (1979) Disruption of maternal behaviorand appearance of cannibalism after ventral mesencephalictegmentum lesions. Physiology and Behavior 23: 317–323.

Garcia-Estrada J, Del Rio JA, Luquin S, Soriano E, and Garcia-Segura LM (1993) Gonadal hormones down-regulatereactive gliosis and astrocyte proliferation after a penetratingbrain injury. Brain Research 628: 271–278.

Garcia-Estrada J, Luguin S, Fernandez M, and Garcia-Segura LM (1999) Dehydroepiandrosterone, pregnanoloneand sex steroids down-regulate reactive astroglia in the malerat brain after a penetrating brain injury. International Journalof Developmental Neuroscience 17: 145–151.

Gatewood JD, Morgan MD, Eaton M, et al. (2005) Motherhoodmitigates aging-related decrements in learning and memory.Brain Research Bulletin 66: 91–98.

Gilbert CL, Burne THJ, Goode JA, Murfitt PJE, and Walton SL(2002) Indomethacin blocks prepartum nest buildingbehaviour in the pig (Sus scrofa): Effects on plasmaprostaglandin F metabolite, OT, cortisol and progesterone.Journal of Endocrinology 172: 507–517.

Giordano AL, Ahdieh HB, Mayer AD, Siegel HI, andRosenblatt JS (1990) Cytosol and nuclear estrogen receptorbinding in the preoptic area and hypothalamus of female ratsduring pregnancy and ovariectomized, nulliparous rats aftersteroid priming: Correlation with maternal behavior.Hormones and Behavior 24: 232–255.

Giordano AL, Siegel HI, and Rosenblatt JS (1989) Nuclearestrogen receptor binding in the preoptic area andhypothalamus of pregnancy-terminated rats:Correlation with the onset of maternal behavior.Neuroendocrinology 50: 248–258.

Giovenardi M, Padoin MJ, Cadore LP, and Lucion AB (1997)Hypothalamic paraventricular nucleus, oxytocin, andmaternal aggression in rats. Annals of the New YorkAcademy of Sciences 807: 606–609.

Giovenardi M, Padoin MJ, Cadore LP, and Lucion AB (1998)Hypothalamic paraventricular nucleus modulates maternalaggression in rats: Effects of ibotenic acid lesion andoxytocin antisense. Physiology and Behavior 63: 351–359.

Gonzalez-Mariscal G (2007) Mother rabbits and their offspring:Timing is everything. Developmental Psychobiology 49:71–76.

Gonzalez-Mariscal G, Chirino R, Beyer C, and Rosenblatt JS(2004) Removal of the accessory olfactory bulbs facilitatesmaternal behavior in virgin rabbits. Behavioural BrainResearch 152: 89–95.

Gonzalez-Mariscal G, Chirino R, Rosenblatt JS, and Beyer C(2005) Forebrain implants of estradiol stimulate maternalnest-building in ovariectomized rabbits. Hormones andBehavior 47: 272–279.

Gonzalez-Mariscal G, Dıaz-Sanchez V, Melo AI, Beyer C, andRosenblatt JS (1994) Maternal behavior in New Zealandwhite rabbits: Quantification of somatic events, motorpatterns, and steroid plasma levels. Physiology and Behavior55: 1081–1089.

Gonzalez-Mariscal G, Flores-Alonso JC, Chirino R,Rosenblatt JS, and Beyer C (2004) Intracerebroventricularinjections of prolactin counteract the antagonistic effect ofbromocriptine on rabbit maternal behaviour. Journal ofNeuroendocrinology 16: 949–955.

Gonzalez-Mariscal G and Gallegos JA (2007) New Zealandwhite rabbits show non-selective nursing in various types ofnests. World Rabbit Science 15: 167–172.

Gonzalez-Mariscal G, Jimenez P, Beyer C, and Rosenblatt JS(2003) Androgens stimulate specific aspects of maternalnest-building and reduce food intake in rabbits. Hormonesand Behavior 43: 312–317.

Gonzalez-Mariscal G, Melo AI, Chirino R, Jimenez P, Beyer C,and Rosenblatt JS (1998) Importance of mother/young contact at parturition and across lactation for theexpression of maternal behavior in rabbits. DevelopmentalPsychobiology 32: 101–111.

Gonzalez-Mariscal G, Melo AI, Parlow AF, Beyer C, andRosenblatt JS (2000) Pharmacological evidence thatprolactin acts from late gestation to promote maternalbehavior in rabbits. Journal of Neuroendocrinology 12:983–992.

Gonzalez-Mariscal G and Poindron P (2002) Parentalcare in mammals: Immediate and sensory factors ofcontrol. In: Pfaff D, Arnold AP, Etgen AM, Fahrbach SE,and Rubin RT (eds.) Hormones, Brain and Behavior,1st edn., vol. 1, ch. 2, pp. 215–298. San Diego, CA:Academic Press.

Gould E, Woolley CS, Frankfurt M, and McEwen BS (1990)Gonadal steroids regulate dendritic spine density inhippocampal pyramidal cells in adulthood. Journal ofNeuroscience 10: 1286–1291.

Graham MD, Rees SL, Steiner M, and Fleming AS (2006) Theeffects of adrenalectomy and corticosterone replacement onmaternal memory in postpartum rats. Hormones andBehavior 49: 353–361.

Gray P and Brooks PJ (1984) Effect of lesion location within themedial preoptic-anterior hypothalamic continuum onmaternal and male sexual behaviors in female rats.Behavioral Neuroscience 98: 703–711.

Gregg JK and Wynne-Edwards KE (2006) In uniparentalPhodopus sungorus new mothers and fathers, presentduring the birth of their offspring, are the only hamsters thatreadily consume fresh placenta. DevelopmentalPsychobiology 48: 528–536.

Hansen S and Ferreira A (1986) Food intake, aggression, andfear behavior in the mother rat: Control by neural systemsconcerned with milk ejection and maternal behavior.Behavioral Neuroscience 100: 64–70.

Hansen S and Gummesson BM (1982) Participation of thelateral midbrain tegmentum in the neuroendocrine controlof sexual behavior and lactation in the rat. Brain Research251: 319–325.

Hansen S and Kohler C (1984) The importance of theperipeduncular nucleus in the neuroendocrine control ofsexual behavior and milk ejection in the rat.Neuroendocrinology 39: 563–572.

Hatton GI and Ellisman MH (1982) A restructuring ofhypothalamic synapses is associated with motherhood.Journal of Neuroscience 2: 704–707.

Hatton GI and Yang QZ (1989) Supraoptic nucleus afferentsfrom the main olfactory bulb – II. Intracellularly recordedresponses to lateral olfactory tract stimulation in rat brainslices. Neuroscience 31: 289–297.

Hatton GI and Yang QZ (1990) Activation of excitatoryaminoacid inputs to supraoptic neurons. I. Induced increasein dye-coupling in lactating, but not virgin or male rats. BrainResearch 513: 264–269.

Hennessy MB (2003) Enduring maternal influences in aprecocial rodent. Developmental Psychobiology 42:225–236.

Herrenkohl LR and Rosenberg PA (1972) Exteroceptivestimulation of maternal behavior in the naive rat. Physiologyand Behavior 8: 595–598.

Herrenkohl LR and Rosenberg PA (1974) Effects ofhypothalamic deafferentation late in gestation onlactation and nursing behavior in the rat. Hormones andBehavior 5: 33–41.

Herrenkohl LR and Sachs BD (1972) Meeting report: Sensoryregulation of maternal behavior in mammals. Physiology andBehavior 9: 689–692.

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 23

HOBB: 00003

Page 24: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Insel TR and Harbaugh CR (1989) Lesions of the hypothalamicparaventricular nucleus disrupt the initiation of maternalbehavior. Physiology and Behavior 45: 1033–1041.

Jacobs LF, Gaulin SJC, Sherry DF, and Hoffman GE (1990)Evolution of spatial cognition: Sex-specific patterns ofspatial behavior predict hippocampal size. Proceedings ofthe National Academy of Sciences of the United States ofAmerica 87: 6349–6352.

Jacobson CD, Terkel J, Gorski RA, and Sawyer CH (1980)Effects of small medial preoptic area lesions on maternalbehavior: Retrieving and nest building in the rat. BrainResearch 194: 471–478.

Jakubowski M and Terkel J (1986) Female reproductive functionand sexually dimorphic prolactin secretion in rats withlesions in the medial preoptic–anterior hypothalamiccontinuum. Neuroendocrinology 43: 696–705.

Jarvis S, Moinard C, Robson SK, et al. (2006) Programmingthe offspring of the pig by prenatal social stress:Neuroendocrine activity and behaviour. Hormones andBehavior 49: 68–80.

Jensen P, Floren K, and Hobroh B (1987) Peri-parturientchanges in behaviour in free-ranging domestic pigs. AppliedAnimal Behaviour Science 17: 69–76.

Johnson MH (2005) Sensitive periods in functional braindevelopment: Problems and prospects. DevelopmentalPsychobiology 46: 287–292.

Kalinichev M, Rosenblatt JS, Nakabeppu Y, and Morrell JI(2000) Induction of c-Fos-like and FosB-likeimmunoreactivity reveals forebrain neuronal populationsinvolved differentially in pup-mediated maternal behavior injuvenile and adult rats. Journal of Comparative Neurology416: 45–78.

Kendrick KM, Da Costa AP, Broad KD, Ohkura S, Guevara R,Levy F, and Keverne EB (1997) Neural control of maternalbehaviour and olfactory recognition of offspring. BrainResearch Bulletin 44: 383–395.

Kendrick KM, Keverne EB, and Baldwin BA (1987)Intracerebroventricular OT stimulates maternal behaviour inthe sheep. Neuroendocrinology 46: 56–61.

Kendrick KM, Levy F, and Keverne EB (1991) Importance ofvaginocervical stimulation for the formation of maternalbonding in primiparous and multiparous parturient ewes.Physiology and Behavior 50: 595–600.

Kendrick KM, Levy F, and Keverne EB (1992) Changes in thesensory processing of olfactory signals induced by birth insheep. Science 256: 833–836.

Kenyon P, Cronin P, and Keeble S (1981) Disruption of maternalretrieving by perioral anesthesia. Physiology and Behavior27: 313–321.

Kenyon P, Cronin P, and Keeble S (1983) Role of the infraorbitalnerve in retreiving behavior in lactating rats. BehavioralNeuroscience 97: 255–269.

Keyser-Marcus L, Stafisso-Sandoz G, Gerecke K, et al. (2001)Alterations of medial preoptic area neurons followingpregnancy and pregnancy-like steroidal treatment in the rat.Brain Research Bulletin 55: 737–745.

Kimchi T, Jennings X, and Dulac C (2007) A functional circuitunderlying male sexual behaviour in the female mouse brain.Nature 448: 1009–1014.

Kinsley CH and Bridges RS (1986) Opiate involvement inpostpartum aggression in rats. Pharmacology, Biochemistry,and Behavior 25: 1007–1011.

Kinsley CH and Bridges RS (1988) Parity associated reductionsin behavioral sensitivity to opiates. Biology of Reproduction39: 270–278.

Kinsley CH and Bridges RS (1990) Morphine and reproductivecondition alter olfactory preferences for pup and adult maleodors in female rats. Developmental Psychobiology 23:331–347.

Kinsley CH and Lambert KG (2006) The maternal brain:Pregnancy and motherhood change the structure of thefemale mammal’s brain making mothers attentive totheir young and better at caring for them. Scientific American294: 72–79.

Kinsley CH, Madonia L, Gifford GW, et al. (1999) Motherhoodimproves learning and memory: Neural activity in rats isenhanced by pregnancy and the demands of rearingoffspring. Nature 402: 137–138.

Kinsley CH, Trainer R, Stafisso-Sandoz G, et al. (2006)Motherhood and pregnancy hormones modifyconcentrations of hippocampal neuronal dendritic spines.Hormones and Behavior 49: 131–142.

Komisaruk BR, Rosenblatt JS, Barona ML, et al. (2000)Combined c-fos and 14C-2-deoxyglucose method todifferentiate site-specific excitation from disinhibition:Analysis of maternal behavior in the rat. Brain Research 859:262–272.

Kozorovitskiy Y, Hughes M, Lee K, and Gould E (2006)Fatherhood affects dendritic spines and vasopressin V1areceptors in the primate prefrontal cortex. NatureNeuroscience 9: 1094–1095.

Krehbiel D, Poindron P, Levy F, and Prud’Homme MJ (1987)Peridural anesthesia disturbs maternal behavior inprimiparous and multiparous parturient ewes. Physiologyand Behavior 40: 463–472.

Kristal MB (1991) Enhancement of opioid-mediated analgesia:A solution to the enigma of placentophagia. Neuroscienceand Biobehavioral Reviews 15: 425–435.

Lambert KG, Berry AE, Griffin G, Amory-Meyers L, Madonia-Lomas L, Love G, and Kinsley CH (2005) Pup exposuredifferentially enhances foraging ability in primiparous andnulliparous rats. Physiology and Behavior 84: 799–806.

Lee A, Clancy S, and Fleming AS (2000) Mother rats bar-pressfor pups: Effects of lesions of the MPOA and limbic sites onmatenal behavior and operant responding for pup-reinforcement. Behavioural Brain Research 108: 215–231.

Levine S (1957) Infantile experience and resistance tophysiological stress. Science 126: 405–406.

Levy F, Keller M, and Poindron P (2004) Olfactory regulationof maternal behavior in mammals. Hormones and Behavior46: 284–302.

Levy F, Kendrick KM, Goode J, Guevara-Guzman R, andKeverne EB (1995) OT and vasopressin release in theolfactory bulb of parturient ewes: Changes with maternalexperience and effects on acetylcholine, gamma-aminobutyric acid, glutamate and noradrenaline release.Brain Research 669: 197–206.

Li C, Brake WG, Romeo RD, et al. (2004) Estrogen altershippocampal dendritic spine shape and enhances synapticprotein immunoreactivity and spatial memory in female mice.Proceedings of the National Academy of Sciences of theUnited States of America 101: 2185–2190.

Li M and Fleming AS (2003) Differential involvement of nucleusaccumbens shell and core subregions in maternal memoryin postpartum female rats. Behavioral Neuroscience 117:426–445.

Lisk RD (1971) Oestrogen and progesterone synergism andelicitation of maternal nest-building in the mouse (Musmusculus). Animal Behaviour 17: 730–738.

Liu Z, Gastard M, Verina T, Bora S, Mouton PR, andKoliatsos VE (2001) Estrogens modulate experimentallyinduced apoptosis of granule cells in the adult hippocampus.Journal of Comparative Neurology 441: 1–8.

Lonstein JS and De Vries GJ (2000) Maternal behaviour inlactating rats stimulates c-fos in glutamate decarboxylase-synthesizing neurons of the medial preoptic area, ventral bednucleus of the stria terminalis, and ventrocaudalperiaqueductal gray. Neuroscience 100: 557–568.

24 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 25: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Lonstein JS, Greco B, De Vries G, Stern JM, and Blaustein JD(2000) Maternal behavior stimulates c-fos activity withinestrogen receptor alpha-containing neurons in lactating rats.Neuroendocrinology 72: 91–101.

Lonstein JS, Simmons DA, Swann JM, and Stern JM (1998)Forebrain expression of c-fos due to active maternalbehaviour in lactating rats. Neuroscience 82: 267–281.

Lonstein JS and Stern JM (1997) Role of the midbrainperiaqueductal gray in maternal nurturance and aggression:c-fos and Electrolytic lesion studies in lactating rats. Journalof Neuroscience 17: 3364–3378.

Lonstein JS and Stern JM (1999) Effects of unilateral suckling onnursing behavior and c-fos activity in the caudalperiaqueductal gray in rats. Developmental Psychobiology35: 264–275.

Love G, Torrey N, McNamara I, et al. (2005) Maternalexperience produces long-lasting behavioral modificationsin the rat. Behavioral Neuroscience 119: 1084–1096.

Lovic V, Gonzalez A, and Fleming AS (2001) Maternallyseparated rats show deficits in maternal care in adulthood.Developmental Psychobiology 39: 19–33.

Macbeth AH, Scharfman HE, MacLusky NJ, Gautreaux C,and Luine VN (2008) Effects of multiparity onrecognition memory, monoaminergic neurotransmitters,and brain-derived neurotrophic factor (BDNF).Hormones and Behavior 54: 7–17.

Maestripieri D, Lindell SG, and Higley JD (2007)Intergenerational transmission of maternal behavior inrhesus macaques and its underlying mechanisms.Developmental Psychobiology 49: 165–171.

Marques DM, Malsbury CW, and Daood J (1979) Hypothalamicknife cuts dissociate maternal behaviors, sexual receptivityand estrous cyclicity in female hamsters. Physiology andBehavior 23: 347–355.

Martin DL and Rimvall K (1993) Regulation of gamma-aminobutyric acid synthesis in the brain. Journal ofNeurochemistry 60: 395–407.

Matthews-Felton T, Corodimas KP, Rosenblatt JS, andMorrell JI (1995) Lateral habenula neurons are necessary forthe hormonal onset of maternal behavior and for the displayof postpartum estrus in naturally parturient female rats.Behavioral Neuroscience 109: 1172–1188.

Matthews-Felton T, Linton LN, Rosenblatt JS, and Morrell JI(1999) Estrogen implants in the lateral habenular nucleus donot stimulate the onset of maternal behavior in female rats.Hormones and Behavior 35: 71–80.

Mayer AD and Rosenblatt JS (1979) Hormonal influences duringthe ontogeny of maternal behavior in female rats. Journal ofComparative and Physiological Psychology 93: 879–898.

McCarthy MM and Konkle ATM (2005) When is a sex differencenot a sex difference? Frontiers in Neuroendocrinology 26:85–102.

McCormack K, Sanchez MM, Bardi M, and Maestripieri D(2006) Maternal care patterns and behavioraldevelopment of rhesus macaque abused infants in thefirst six months of life. Developmental Psychobiology 48:537–550.

Melo AI and Gonzalez-Mariscal G (2003) Placentophagia inrabbits: Incidence across the reproductive cycle.Developmental Psychobiology 43: 37–43.

Melo AI, Lovic V, Gonzalez A, Madden M, Sinopoli K, andFleming AS (2006) Maternal and littermate deprivationdisrupts social behavior and social learning of foodpreference in adulthood: Tactile stimulation, nest odor,and social rearing prevent these effects. DevelopmentalPsychobiology 48: 197–208.

Mena F and Grosvenor CE (1972) Effects of suckling andexteroceptive stimulation upon prolactin release in the ratduring late lactation. Journal of Endocrinology 52: 11–22.

Miceli MO, Fleming AS, and Malsbury CW (1983) Disruption ofmaternal behaviour in virgin and postparturient rats followingsagittal plane knife cuts in the preoptic area–hypothalamus.Behavioural Brain Research 9: 337–360.

Miceli MO and Malsbury CW (1982) Sagittal knife cuts in thenear and far lateral preoptic area–hypothalamus disruptmaternal behavior in female hamsters. Physiology andBehavior 28: 857–867.

Michel GF and Tyler AN (2005) Critical period: A history of thetransition from questions of when, to what, to how.Developmental Psychobiology 46: 156–162.

Miller S and Erskine MS (1995) Ultrastructural effects ofestradiol and 5-alpha-androstane on neurons within theventromedial nucleus of the hypothalamus.Neuroendocrinology 61: 669–679.

Moberg GP and Mench JA (eds.) (2001) The Biology of AnimalStress. New York: CABI Publishing.

Modney BK and Hatton GI (1990) Motherhood modifiesmagnocellular neuronal relationships in functionallymeaningful ways. In: Krasnegor NA and Bridges RS (eds.)Mammalian Parenting: Biochemical, Neurobiological, andBehavioral Determinants, pp. 305–323. New York: OxfordUniversity Press.

Moore CL (1995) Maternal contributions to mammalianreproductive development and the divergence of males andfemales. In: Slater JB, Rosenblatt JS, Snowdon CT, andMilinski M (eds.) Advances in the Study of Behavior, vol. 24,pp. 47–118. San Diego, CA: Academic Press.

Morgan HD, Watchus JA, Milgram NW, and Fleming AS(1999) The long lasting effects of electrical simulation ofthe medial preoptic area and medial amygdala on maternalbehavior in female rats. Behavioural Brain Research 99:61–73.

Morgan JI and Curran T (1991) Stimulus–transcription couplingin the nervous system: Involvement of the inducibleproto-oncogenes fos and jun. Annual Review ofNeuroscience 14: 421–451.

Muccioli G and Di Carlo R (1994) Modulation of prolactinreceptors in the rat hypothalamus in response to changes inserum concentration of endogenous prolactin or to ovineprolactin administration. Brain Research 663: 244–250.

Murphy MR, MacLean PD, and Hamilton SC (1981)Species-typical behavior of hamsters deprived from birthof the neocortex. Science 213: 459–461.

Page 26: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Neumann ID, Torner L, and Wigger A (2000) Brain oxytocin:Differential inhibition of neuroendocrine stress responsesand anxiety-related behaviour in virgin, pregnant andlactating rats. Neuroscience 95: 567–575.

Nowak R, Keller M, Val-Laillet D, and Levy F (2007) Perinatalvisceral events and brain mechanisms involved in thedevelopment of mother–young bonding in sheep. Hormonesand Behavior 52: 92–98.

Nudo RJ and Masterton RB (1986) Stimulation-induced 14C-2-deoxyglucose labeling of synaptic activity in the centralauditory system. Journal of Comparative Neurology 245:553–565.

Numan M (1974) Medial preoptic area and maternal behavior inthe female rat. Journal of Comparative and PhysiologicalPsychology 87: 746–759.

Numan M (1978) Progesterone inhibition of maternal behavior inthe rat. Hormones and Behavior 11: 209–231.

Numan M (1990) Long-term effects of preoptic area knife cutson the maternal behavior of postpartum rats. Behavioraland Neural Biology 53: 284–290.

Numan M (1996) A lesion and neuroanatomical tract-tracinganalysis of the role of the bed nucleus of the striaterminalis in retrieval behavior and other aspects ofmaternal responsiveness in rats. DevelopmentalPsychobiology 29: 23–51.

Numan M and Callahan EC (1980) The connections of themedial preoptic region and maternal behavior in the rat.Physiology and Behavior 25: 653–665.

Numan M and Corodimas KP (1985) The effects ofparaventricular hypothalamic lesions on maternal behavior inrats. Physiology and Behavior 35: 417–425.

NumanM, Corodimas KP, NumanMJ, Factor EM, and PiersWD(1988) Axon-sparing lesions of the preoptic region andsubstantia innominata disrupt maternal behavior in rats.Behavioral Neuroscience 102: 381–396.

Numan M, Fleming AS, and Levy F (2006) The physiology ofreproduction: Maternal behavior. In: Neill JD (ed.) Knobil andNeill’s Physiology of Reproduction, 3rd edn., vol. 2, ch. 35,pp. 1921–1923. San Diego, CA: Elsevier.

Numan M and Insel TR (2003) The Neurobiology of ParentalBehavior. New York: Springer.

Numan M, Morrell JI, and Pfaff DW (1985) Anatomicalidentification of neurons in selected brain regions associatedwith maternal behavior deficits induced by knife cuts of thelateral hypothalamus in rats. Journal of ComparativeNeurology 237: 552–564.

Numan M and Numan MJ (1991) Preoptic–brainstemconnections and maternal behavior in rats. BehavioralNeuroscience 105: 1013–1029.

Numan M and Numan MJ (1994) Expression of Fos-likeimmunoreactivity in the preoptic area of maternally behavingvirgin and postpartum rats. Behavioral Neuroscience 108:379–394.

Numan M and Numan MJ (1995) Importance of pup-relatedsensory inputs and maternal performance for theexpression of fos-like immunoreactivity in the preopticarea and ventral bed nucleus of the stria terminalisof postpartum rats. Behavioral Neuroscience 109: 135–149.

Numan M, Numan MJ, and English JB (1993) Excitotoxicamino acid injections into the medial amygdala facilitatematernal behavior in virgin female rats. Hormones andBehavior 27: 56–81.

Numan M, Numan MJ, Marzella SR, and Palumbo A (1998)Expression of c-fos, fos B, and egr-1 in the medialpreoptic area and bed nucleus of the stria terminalisduring maternal behavior in rats. Brain Research 792:348–352.

Numan M, Roach JK, Del Cerro MC, Guillamon A, Segovia S,Sheehan TP, and Numan MJ (1999) Expression of

intracellular progesterone receptors in rat brain duringdifferent reproductive states, and involvement in maternalbehavior. Brain Research 830: 358–371.

Numan M, Rosenblatt JS, and Komisaruk BR (1977) Medialpreoptic area and onset of maternal behavior in the rat.Journal of Comparative and Physiological Psychology 91:146–164.

Numan M and Smith HG (1984) Maternal behavior in rats:Evidence for the involvement of preoptic projections to theventral tegmental area. Behavioral Neuroscience 98:712–727.

Olazabal DE and Ferreira A (1997) Maternal behavior in rats withkainic acid-induced lesions of the hypothalamicparaventricular nucleus. Physiology and Behavior 61:779–784.

Parrish JZ, Emoto K, Kim MD, and Jan YN (2007) Mechanismsthat regulate establishment maintenance and remodelingof dendritic fields. Annual Review of Neuroscience 30:399–423.

Pawluski JL and Galea LAM (2005) Hippocampalmorphology is differentially affected byreproductive experience in the mother. Journal ofNeurobiology 66: 71–81.

Pawluski JL, Vanderbyl BL, Ragan K, and Galea LAM (2006a)First reproductive experience persistently affects spatialreference and working memory in the mother and theseeffects are not due to pregnancy or mothering alone.Behavioural Brain Research 175: 157–165.

Pawluski JL, Walker SK, and Galea LAM (2006b) Reproductiveexperience differentially affects spatial reference andworking memory performance in the mother. Hormones andBehavior 49: 143–149.

Pedersen CA, Caldwell JD, Johnson MF, Fort SA, andPrange AJJ (1985) OT antiserum delays onset of ovariansteroid-induced maternal behavior. Neuropeptides 6:175–182.

Pereira M and Ferreira A (2006) Demanding pups improvematernal behavioral impairments in sensitized andhaloperidol-treated lactating female rats. Behavioural BrainResearch 175: 139–148.

Perlmutter LS, Tweedle CD, and Hatton GI (1984) Neuronal–glialplasticity in the supraoptic dendritic zone: Dendritic bundlingand double synapse formation at parturition. Neuroscience13: 769–779.

Pfaff DW and Keiner M (1973) Atlas of estradiol-concentratingcells in the central nervous system of the rat. Journal ofComparative Neurology 141: 121–158.

Pi XJ and Grattan DR (1998) Distribution of prolactinreceptor immunoreactivity in the brain of estrogen-treated,ovariectomized rats. Journal of Comparative Neurology394: 462–474.

Pi X and Grattan DR (1999a) Expression of prolactin receptormRNA is increased in the preoptic area of lactating rats.Endocrine 11: 91–98.

Pi XJ and Grattan DR (1999b) Increased expression of bothshort and long forms of prolactin receptor mRNA inhypothalamic nuclei of lactating rats. Journal of MolecularEndocrinology 23: 13–22.

Pi XJ and Grattan DR (1999) Increased prolactin receptorimmunoreactivity in the hypothalamus of lactating rats.Journal of Neuroendocrinology 11: 693–705.

Poindron P, Levy F, and Keller M (2007a) Maternalresponsiveness and maternal selectivity in domestic sheepand goats: The two facets of maternal attachment.Developmental Psychobiology 49: 54–70.

Poindron P, Terrazas A, Navarro Montes de Oca ML, Serafın N,and Hernandez H (2007b) Sensory and physiologicaldeterminants of maternal behavior in the goat (Capra hircus).Hormones and Behavior 52: 99–105.

26 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 27: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Rasia-Filho AA, Fabian C, Rigoti KM, and Achaval M (2004)Influence of sex estrous cycle and motherhood on dendriticspine density in the rat medial amygdala revealed by theGolgi method. Neuroscience 126: 839–847.

Rees SL and Fleming AS (2001) The effects of early separationand juvenile exposure to pups on maternal behavior in adultpostpartum rats. Animal Learning and Behavior 29: 221–233.

Rosenblatt JS and Lehrman DS (1963) Maternal behavior in thelaboratory rat. In: Rheingold HL (ed.) Maternal Behavior inMammals, pp. 8–57. New York: Wiley.

Salm AK, Modney BK, and Hatton GI (1988) Alterations insupraoptic nucleus ultrastructure of maternally behavingvirgin rats. Brain Research Bulletin 21: 685–691.

Scharfman HE, Mercurio TC, Goodman JH, Wilson MA, andMacLusky N (2003) Hippocampal excitability increasesduring the estrous cycle in the rat: A potential role forbrain-derived neurotrophic factor. Journal of Neuroscience17: 11641–11652.

Scott CJ, Pereira AM, Rawson JA, Simmons DM,Rossmanith WG, Ing NH, and Clark IJ (2000) The distributionof progesterone receptor immunoreactivity in the preopticarea and hypothalamus of the ewe: Upregulation ofprogesterone receptor mRNA in the mediobasalhypothalamus by oestrogen. Journal of Neuroendocrinology12: 565–575.

Sharp JS, Gonzalez MF, Morton MT, Simon RP, and Sharp FR(1988) Decreases of cortical and thalamic glucosemetabolism produced by parietal cortex stimulation in therat. Brain Research 438: 357–362.

Sheehan TP, Cirrito J, Numan MJ, and Numan M (2000) Usingc-Fos immunocytochemistry to identify forebrain regionsthat may inhibit maternal behavior in rats. BehavioralNeuroscience 114: 337–352.

Slotnick BM and Nigrosh BJ (1975) Maternal behavior ofmice with cingulate cortical, amygdala, or septal lesions.Journal of Comparative and Physiological Psychology 88:118–127.

Smith RL (1973) The ascending fibers from the principaltrigeminal sensory nucleus in the rat. Journal of ComparativeNeurology 148: 423–446.

Solum DT and Handa RJ (2002) Estrogen regulates thedevelopment of brain-derived neurotrophic factor mRNAand protein in the rat hippocampus. Journal of Neuroscience22: 2650–2659.

Stack EC and Numan M (2000) The temporal course ofexpression of c-Fos and FosB within the medial preopticarea and other brain regions of postpartum female ratsduring prolonged mother–young interactions. BehavioralNeuroscience 114: 609–622.

Stafisso-Sandoz G, Polley G, Holt E, Lambert KG, andKinsley CH (1998) Opiate disruption of maternal behavior:Morphine reduces and naloxone restores c-fos activity in themedial preoptic area of lactating rats. Brain ResearchBulletin 45: 307–313.

Stern JM (1977) Effects of ergocryptine on postpartummaternalbehavior, ovarian cyclicity, and food intake in rats.Behavioral Biology 21: 134–140.

Stern J (1996) Somatosensation and maternal care inNorway rats. In: Rosenblatt JS and Snowdon CT (eds.)Parental Care: Evolution, Mechanisms and AdaptiveSignificance, vol. 25, pp. 243–294. New York: AcademicPress.

Stern JM, Dix L, Bellomo C, and Thramann C (1992) Ventraltrunk somatosensory determinants of nursing behavior inNorway rats. II. Role of nipple and surrounding sensations.Psychobiology 20: 71–80.

Stern JM and Johnson SK (1990) Ventral somatosensorydeterminants of nursing behavior in Norway rats (Rattusnorvegicus). Physiology and Behavior 47: 993–1011.

Stern JM and Kolunie JM (1989) Perioral anesthesia disruptsmaternal behavior during early lactation in Long-Evans rats.Behavioral and Neural Biology 52: 20–38.

Swanson LJ and Campbell CS (1979) Induction of maternalbehavior in nulliparous golden hamsters (Mesocricetusauratus). Behavioral and Neural Biology 26: 364–371.

Swanson LW (1976) An autoradiographic study of the efferentconnections of the preoptic region in the rat. Journal ofComparative Neurology 167: 227–256.

Szyf M, Weaver ICG, Champagne FA, Diorio J, and Meaney MJ(2005) Maternal programming of steroid receptorexpression and phenotype through DNA methylationin the rat. Frontiers in Neuroendocrinology 26: 139–162.

Terkel J, Bridges RS, and Sawyer CH (1979) Effects oftransecting lateral neural connections of the medial preopticarea on maternal behavior in the rat: Nest building, pupretrieval and prolactin secretion. Brain Research 169:369–380.

Terlecki LJ and Sainsbury RS (1978) Effects of fimbria lesions onmaternal behavior in the rat. Physiology and Behavior 21:89–97.

Theodosis DT, Montagnese C, Rodriguez F, Vincent JD, andPoulain DA (1986) Oxytocin induces morphological plasticityin the adult hypothalamo-neurohypophysial system. Nature322: 738–774.

Theodosis DT and Poulain DT (1984) Evidence for structuralplasticity in the supraoptic nucleus of the rat hypothalamusin relation to gestation and lactation. Neuroscience 11:183–193.

Theodosis DT, Poulain DA, and Vincent JD (1981) Possiblemorphological bases of synchronisation of neuronal firingin the rat supraoptic nucleus during lactation. Neuroscience6: 919–929.

Tomizawa K, Iga N, Lu Y, et al. (2003) Oxytocin improveslong-lasting spatial memory during motherhoodthrough MAP kinase cascade. Nature Neuroscience 6:384–390.

Van Leengoed E, Kerker E, and Swanson HJ (1987) Inhibition ofpostparum maternal behavior in the rat by injecting an OTantagonist into the cerebral ventricles. Journal ofEndocrinology 112: 275–282.

Vermetten E and Bremner JD (2002) Circuits and systems instress. Depression and Anxiety 15: 126–147.

Wagner CK and Morrell JI (1996) Levels of estrogen receptorimmunoreactivity are altered in behaviorally-relevantbrain regions in female rats during pregnancy. BrainResearch – Molecular Brain Research 42: 328–336.

Wagner CK, Silverman AJ, and Morrell JI (1998) Evidence forestrogen receptor in cell nuclei and axon terminalswithin the lateral habenula of the rat: Regulationduring pregnancy. Journal of Comparative Neurology 392:330–342.

Walsh CJ, Fleming AS, Lee A, and Magnusson JE (1996) Theeffects of olfactory and somatosensory desensitization onFos-like immunoreactivity in the brains of pup-exposedpostpartum rats. Behavioral Neuroscience 110: 134–153.

Walsh RJ, Mangurian LP, and Posner BI (1990) The distributionof lactogen receptors in the mammalian hypothalamus: Anin vitro autoradiographic analysis of the rabbit and rat. BrainResearch 530: 1–11.

Warembourg M, Jolivet A, and Milgrom E (1989)Immunohistochemical evidence of the presence ofestrogen and progesterone receptors in the sameneurons of the guinea pig hypothalamus and preopticarea. Brain Research 480: 1–15.

Warembourg M, Logeat F, and Milgrom E (1986)Immunocytochemical localization of progesteronereceptor in the guinea pig central nervous system.Brain Research 384: 121–131.

From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain 27

HOBB: 00003

Page 28: HOBB: 00003 - Elsevier

ELSE

VIER

THIRDPR

OOF

Warren SG, Humphreys AG, Juraska JM, and Greenough WT(1995) LTP varies across the estrus cycle: Enhanced synapticplasticity in proestrous rats. Brain Research 703: 26–30.

Warren SG and Juraska JM (1997) Spatial and non-spatiallearning across the rat estrous cycle. BehavioralNeuroscience 111: 259–266.

Wartella J, Amory E, Madonia-Lomas L, et al. (2003) Single ormultiple reproductive experiences attenuateneurobehavioral stress and fear responses in the female rat.Physiology and Behavior 79: 373–381.

Weaver ECG, Champagne FA, Brown SE, Dymov S, Sharma S,Meaney MJ, and Szyf M (2005) Reversal of maternalprogramming of stress responses in adult offspring throughmethyl supplementation: Altering epigenetic marking later inlife. Journal of Neuroscience 25: 11045–11054.

Weaver ICG, Cervoni N, Champagne FA, et al. (2004) Epigeneticprogramming by maternal behavior. Nature Neuroscience7: 847–854.

Wewers D, Kaiser S, and Sachser N (2003) Maternal separationin guinea pigs: A study in behavioural endocrinology.Ethology 109: 443–453.

Wisden W, Errington ML, Williams S, et al. (1990) Differentialexpression of immediate early genes in the hippocampusand spinal cord. Neuron 4: 603–614.

Woolley CS (1998) Estrogen-mediated structural and functionalsynaptic plasticity in the female rat hippocampus. Hormonesand Behavior 34: 140–148.

Woolley CS, Gould E, Frankfurt M, and McEwen BS (1990)Naturally occurring fluctuations in dendritic spine density onadult hippocampal pyramidal neurons. Journal ofNeuroscience 10: 4035–4039.

Woolley CS and McEwen BS (1992) Estradiol mediatesfluctuation in hippocampal synapse density during the estrouscycle in the adult. Journal of Neuroscience 12: 2549–2554.

Woolley CS and McEwen BS (1993) Roles of estradiol andprogesterone in the regulation of hippocampal dendriticspine density during the estrous cycle in the rat. Journal ofComparative Neurology 336: 293–306.

Woolley CS, Wenzel HJ, and Schwartzkroin PA (1996)Estradiol increases the frequency of multiple synapseboutons in the hippocampal CA1 region of the adult femalerat. Journal of Comparative Neurology 373: 108–117.

Xerri C, Stern JM, and Merzenich M (1994) Alterations of thecortical representation of the rat ventrum induced by nursingbehavior. Journal of Neuroscience 14: 1710–1721.

Yankova M, Hart SA, andWoolley CS (2001) Estrogen increasessynaptic connectivity between single presynaptic inputsand multiple postsynaptic CA1 pyramidal cells: A serialelectron-microscopic study. Proceedings of the NationalAcademy of Sciences of the United States of America 98:3525–3530.

Zarrow MX, Denenberg VH, and Anderson CO (1965)Rabbit: Frequency of suckling in the pup. Science 150:1835–1836.

Further Reading

Gonzalez A, Lovic V, Ward GR, Wainwright PE, and Fleming AS(2001) Intergenerational effects of complete maternaldeprivation and replacement stimulation on maternalbehavior and emotionality in female rats. DevelopmentalPsychobiology 38: 11–32.

Modney BK and Hatton GI (1994) Maternal behaviors: Evidencethat they feed back to alter brain morphology and function.Acta Paediatrica 397(supplement): 29–32.

28 From Indifference to Ardor: The Onset, Maintenance, and Meaning of the Maternal Brain

HOBB: 00003

Page 29: HOBB: 00003 - Elsevier

Non-Print Items

Abstract:

We describe and review maternal behavior in mammals (viz., rodents, rabbits, and ungulates), and theparticipation of steroid/peptide hormones in its regulation (e.g., nest-building, nursing, and aggres-sion). We discuss the neurobiology involved in the onset and offset of specific behavioral patterns andthe transmission and interpretation of signals from the young, which facilitate the offspring’s devel-opment. The impact of mothering styles on the psychobiological development of organisms and theiremotional reactivity as adults is addressed, as well as the epigenetic mechanisms participating in theintergenerational transmission of such acquired traits. We also discuss the ancillary and complemen-tary changes that occur to the mother’s brain and behavior, which support the display of care andprotection for young, and which extend to well beyond the peripartum and lactation periods of themother’s life. Such behavioral alterations, among them enhanced cognition and reductions in fear andanxiety, translate into more efficient behavior and a cost:benefit ratio that favors the survival of bothmother and young. We draw attention to topics that merit further investigation (e.g., epigenetictransmission of behavior in mammals other than rodents and mechanisms involved in the facilitationof cognition by motherhood) and to little explored issues (e.g., placentophagia, nest-building, and foodpreferences).

Keywords: Amygdala; Anxiety; BDNF; Epigenetics; Hippocampus; Lactation; Mammals; Medialpreoptic area; Neuroplasticity; Parity; Parturition; Postpartum; Prepartum; Pup sensitization; Spatialmemory; Stress

Author and Co-author Contact Information:

Gabriela Gonzalez-MariscalCINVESTAV – Universidad Autonoma de TlaxcalaApdo Postal 62TlaxcalaTlax [email protected]

Craig Howard KinsleyDepartment of Psychology-Center for NeuroscienceGottwald Science CenterUniversity of Richmond28Westhampton WayRichmondVA [email protected]

HOBB: 00003

Page 30: HOBB: 00003 - Elsevier
Page 31: HOBB: 00003 - Elsevier

(1979) and a PhD from the State University of New York, University Center at Albany (1985). From 1985 to1989, he was a postdoctoral fellow and research associate in the Laboratory of Human Reproduction and

Reproductive Biology, Department of Anatomy, at Harvard Medical School, and an instructor at HarvardUniversity. He joined the University of Richmond and its Department of Psychology in 1989. He and colleagueKelly Lambert recently published the textbook, Clinical Neuroscience: The Neurobiological Foundations of Mental

Health (1st edition: Worth, 2006; 2nd edition: Oxford University Press, projected, 2009). His research is funded by

the US National Institutes of Health, the National Science Foundation, and by private foundations such as theHoward Hughes Medical Institute, the Keck Foundation, and Arnold O. Beckman Foundation. Among hisachievements since his arrival at Richmond, he is most proud of the 100+ students he has trained who have

matriculated to PhD, MD, and MA programs in neuro-related areas.

HOBB: 00003