Elsevier

Hormones and Behavior

Volume 62, Issue 2, July 2012, Pages 128-135
Hormones and Behavior

Experience-facilitated improvements in pup retrieval; evidence for an epigenetic effect

https://doi.org/10.1016/j.yhbeh.2012.05.012Get rights and content

Abstract

The quality and quantity of maternal care received during infancy are highly predictive of successful infant development. It has been well established, primarily in rats, that the combination of hormonal and infant stimuli at birth modifies neural circuits that regulate maternal responsiveness. During subsequent interactions, infant stimuli are more likely to elicit rapid maternal responsiveness. Some species, such as humans, can display maternal care in the absence of the endocrine events of pregnancy and birth. Similarly, virgin C57BL/6J female mice, display maternal care toward infants, and experience with infants elicits long-lasting increases in maternal care. We hypothesized that these experience-induced changes in behavior may be mediated by chromatin modifications, which in turn change expression of genes that promote maternal care. One site of action is the medial preoptic area (MPOA). To test our hypothesis we treated virgin female mice with sodium butyrate, a histone deacetylase inhibitor. This treatment potentiated maternal responsiveness as well as the expression of several genes: estrogen receptor β (Esr2), oxytocin (Oxt), and cyclicAMP response element binding protein (CREB) binding protein (Crebbp; a histone acetyltransferase) in the MPOA. These data suggest that experience induces high levels of maternal care via epigenetic modifications.

Highlights

► Maternal experience facilitates subsequent maternal care in C57/BL6J mice. ► Experience-dependent effects are associated with increased gene expression in medial preoptic area. ► A histone deacetylase inhibitor potentiated experience effects on behavior and gene expression.

Introduction

In humans and other mammals, experience with infants has substantial effects on the quality of subsequent maternal care, which in turn affects infant development. In human mothers, more contact with infants during the first hours–days postpartum is associated with increased maternal responding, reduced infant crying, and an increase in secure infant attachment (Bystrova et al., 2009, Erlandsson et al., 2007, Kennell and Klaus, 1998, Klaus et al., 1972). For at risk mothers, greater infant contact is significantly correlated with a decreased incidence of child abuse/neglect (Buranasin, 1991, O'Connor et al., 1980). Therefore, understanding how experience with infants can produce changes in maternal care is essential for understanding how these mechanisms might fail in mothers that fail to bond with their infants.

The mechanisms through which mother–infant interactions act on the brain to alter subsequent maternal responsiveness have been best characterized in rats, in which the combination of hormonal and infant stimuli at birth permanently enhances maternal responsiveness (Fleming and Korsmit, 1996, Fleming et al., 1999, Numan, 2006). For example, postpartum rats are highly responsive to pups and will learn to press a lever or traverse a novel environment to retrieve pups back to the nest (Fleming et al., 1994, Lee et al., 2000, Stern and Mackinnon, 1976). Even in the absence of continued hormone or infant exposure, female rats show long-lasting changes in maternal responsiveness (Bridges, 1975, Bridges, 1977, Bridges, 1978, Orpen and Fleming, 1987, Orpen et al., 1987, Scanlan et al., 2006).

The medial preoptic area (MPOA), is the critical neural site that responds to both hormonal and sensory inputs from pups and regulates behavior (Arrati et al., 2006, Fleming et al., 1983, Gray and Brooks, 1984, Jacobson et al., 1980, Kalinichev et al., 2000, Lee and Brown, 2007, Lee et al., 2000, Numan, 1974, Numan and Callahan, 1980, Numan et al., 1977, Numan et al., 1988). The MPOA undergoes a variety of changes pre- versus post-partum that promote maternal responsiveness (Afonso et al., 2009, Febo et al., 2005, Fleming and Korsmit, 1996, Kim et al., 2010, Kuroda et al., 2007, Meddle et al., 2007, Numan and Numan, 1994, Numan and Numan, 1995, Numan and Numan, 1997, Numan et al., 1998, Seifritz et al., 2003, Stack et al., 2002). Thus, interaction with infants in the context of these changes, likely modifies the MPOA such that during subsequent interactions, infant stimuli come to elicit maternal responsiveness more effectively.

However, not all species rely on the hormonal stimulation of birth to respond to infants. For example, when foster pups are scattered in the home cage of virgin laboratory mice, they respond to pups (retrieve, lick, crouch) within 15 min (Calamandrei and Keverne, 1994, Gandelman, 1973a, Gandelman, 1973b, Gandelman and Vom Saal, 1975, Kuroda et al., 2008, Larsen et al., 2008, Leussis et al., 2008, Lucas et al., 1998, Mann et al., 1983, Noirot, 1972, Okabe et al., 2011, Stolzenberg and Rissman, 2011, Thomas and Palmiter, 1997). We have recently shown that experience with pups can initiate and sustain maternal responsiveness in a novel T-maze (high levels of maternal responsiveness) in virgin C57BL6/J mice. Moreover, behaviors displayed by virgins are not significantly different from postpartum females (Stolzenberg and Rissman, 2011). In addition, only 4 days of experience (for just 2 h/day) with pups is required for females to show high levels of maternal responsiveness, whereas mice with 50% less pup experience (2 h/day for 2 days) do not show this experience-induced increase in high levels of maternal responsiveness.

A critical question then is how this subtle difference in maternal experience can induce high levels of maternal responsiveness. We hypothesize that, in the absence of pregnancy and parturition, multiple experiences with infants may allow the transcription of genes that are typically regulated by the combination of hormonal stimulation and mother–infant interaction at birth to be activated in virgin mice. We speculated that epigenetic control of gene expression might contribute to the acute regulation of gene expression in response to experiences (Sweatt, 2009). One mechanism through which experience-dependent behavioral modifications are consolidated is epigenetic histone acetylation. Addition of acetyl groups, by histone acetyltransferases (HATs), to the histone proteins around which DNA is wrapped increases the sensitivity of DNA to transcriptional regulation.

In order to explore these hypotheses, we used a histone deacetylase inhibitor (HDAC inhibitor), sodium butyrate (SB), which inhibits HDAC activity and increases histone acetylation (Roozendaal et al., 2010). In the first experiment we examined whether HDAC inhibition would potentiate the effects of maternal experience on subsequent maternal behavior. In experiments 2 and 3 we examined the specificity of SB effects on maternal responsiveness. Finally, as a first step toward addressing how HDAC inhibition might mediate effects, we asked whether SB upregulated the expression of genes that are known to be associated with maternal experience.

Section snippets

Subjects and drug treatment

All mice were C57BL/6J virgin nulliparous females (60–100 days of age), naive to pups (except for their own littermates). Sodium butyrate (SB; Sigma-Aldrich, MO) was dissolved in sterile water. The drug was diluted to a dose of 8 mg/ml in the drinking water. Control mice received the vehicle (water) in the drinking water. Oral administration of SB at this dose increases histone acetylation in the hypothalamus (including MPOA) (Bonthuis et al., 2011). SB treatment began 10 days prior to testing and

Experiment 1: effects of HDAC inhibition on maternal experience-dependent maternal responsiveness

Upon initial exposure to pups in the home cage, SB treated females were significantly faster to retrieve all pups to the nest [main effect of Treatment F(1,12) = 6.81, P = 0.02] when compared with Water treated control females (Table 1). Post hoc analyses revealed that SB treated females were significantly different than Water treated females on Test Day 1. Although there were no significant effects of SB treatment on latency to group all pups inside the nest [F(1,12) = 2.67, P = 0.13], or crouch over

Discussion

Here we report that treatment with SB, an HDAC inhibitor, amplified the effects of maternal experience, both on maternal responsiveness in a novel environment and on gene expression in the MPOA. We have previously reported that virgin female mice require 4 days (2 h/day) of maternal experience in order to show high levels of maternal responsiveness on a novel T-maze (Stolzenberg and Rissman, 2011). Thus, treatment with SB effectively reduced by 50% the amount of maternal experience required to

Acknowledgments

The authors thank A. Ryalls, S. Shetty, and M. Edwards for their outstanding technical assistance. This work has been supported by the National Institutes of Health T32 DK007646 and R01 MH057759. The University of Virginia Center for Research in Reproduction Ligand Assay and Analysis Core is supported by the Eunice Kennedy Shriver NICHD/NIH (SCCPIR) Grant U54-HD28934.

References (102)

  • A.S. Fleming et al.

    Lesions of the medial preoptic area prevent the facilitation of maternal behavior produced by amygdala lesions

    Physiol. Behav.

    (1983)
  • A.S. Fleming et al.

    Neurobiology of mother–infant interactions: experience and central nervous system plasticity across development and generations

    Neurosci. Biobehav. Rev.

    (1999)
  • S.C. Gammie et al.

    Gene array profiling of large hypothalamic CNS regions in lactating and randomly cycling virgin mice

    Brain Res. Mol. Brain Res.

    (2005)
  • R. Gandelman

    Maternal behavior in the mouse: effect of estrogen and progesterone

    Physiol. Behav.

    (1973)
  • R. Gandelman

    The ontogeny of maternal responsiveness in female Rockland-Swiss albino mice

    Horm. Behav.

    (1973)
  • R. Gandelman et al.

    Pup-killing in mice: the effects of gonadectomy and testosterone administration

    Physiol. Behav.

    (1975)
  • B.B. Gundersen et al.

    Effects of the histone deacetylase inhibitor sodium butyrate in models of depression and anxiety

    Neuropharmacology

    (2009)
  • C.D. Jacobson et al.

    Effects of small medial preoptic area lesions on maternal behavior: retrieving and nest building in the rat

    Brain Res.

    (1980)
  • S.H. Jin et al.

    Cyclic AMP response element-binding protein is required for normal maternal nurturing behavior

    Neuroscience

    (2005)
  • K.M. Kendrick et al.

    Neural control of maternal behaviour and olfactory recognition of offspring

    Brain Res. Bull.

    (1997)
  • E. Korzus et al.

    CBP histone acetyltransferase activity is a critical component of memory consolidation

    Neuron

    (2004)
  • K.O. Kuroda et al.

    ERK-FosB signaling in dorsal MPOA neurons plays a major role in the initiation of parental behavior in mice

    Mol. Cell. Neurosci.

    (2007)
  • K.O. Kuroda et al.

    Neurobehavioral basis of the impaired nurturing in mice lacking the immediate early gene FosB

    Brain Res.

    (2008)
  • C.M. Larsen et al.

    Male pheromones initiate prolactin-induced neurogenesis and advance maternal behavior in female mice

    Horm. Behav.

    (2008)
  • A.W. Lee et al.

    Comparison of medial preoptic, amygdala, and nucleus accumbens lesions on parental behavior in California mice (Peromyscus californicus)

    Physiol. Behav.

    (2007)
  • M.P. Leussis et al.

    Attenuation of maternal behavior in virgin CD-1 mice by methylphenidate hydrochloride

    Physiol. Behav.

    (2008)
  • J.M. Levenson et al.

    Regulation of histone acetylation during memory formation in the hippocampus

    J. Biol. Chem.

    (2004)
  • M. Malvaez et al.

    Modulation of chromatin modification facilitates extinction of cocaine-induced conditioned place preference

    Biol. Psychiatry

    (2010)
  • M.A. Mann et al.

    Infanticide exhibited by female mice: genetic, developmental and hormonal influences

    Physiol. Behav.

    (1983)
  • K.R. Mifsud et al.

    Epigenetic mechanisms in stress and adaptation

    Brain Behav. Immun.

    (2011)
  • E. Noirot

    The onset of maternal behavior in rat, hamsters and mice

  • M. Numan et al.

    The connections of the medial preoptic region and maternal behavior in the rat

    Physiol. Behav.

    (1980)
  • M. Numan et al.

    Expression of c-fos, fos B, and egr-1 in the medial preoptic area and bed nucleus of the stria terminalis during maternal behavior in rats

    Brain Res.

    (1998)
  • B.G. Orpen et al.

    Experience with pups sustains maternal responding in postpartum rats

    Physiol. Behav.

    (1987)
  • B.G. Orpen et al.

    Hormonal influences on the duration of postpartum maternal responsiveness in the rat

    Physiol. Behav.

    (1987)
  • E. Seifritz et al.

    Sustained blood oxygenation and volume response to repetition rate-modulated sound in human auditory cortex

    NeuroImage

    (2003)
  • E.C. Stack et al.

    A functional neuroanatomical investigation of the role of the medial preoptic area in neural circuits regulating maternal behavior

    Behav. Brain Res.

    (2002)
  • J.M. Stern et al.

    Postpartum, hormonal, and nonhormonal induction of maternal behavior in rats: effects on T-maze retrieval of pups

    Horm. Behav.

    (1976)
  • J.D. Sweatt

    Experience-dependent epigenetic modifications in the central nervous system

    Biol. Psychiatry

    (2009)
  • S.A. Thomas et al.

    Impaired maternal behavior in mice lacking norepinephrine and epinephrine

    Cell

    (1997)
  • D. Benoit et al.

    Stability and transmission of attachment across three generations

    Child Dev.

    (1994)
  • P.J. Bonthuis et al.

    Acquisition of sexual receptivity: roles of chromatin acetylation, estrogen receptor-alpha, and ovarian hormones

    Endocrinology

    (2011)
  • O.J. Bosch et al.

    Maternal behaviour is associated with vasopressin release in the medial preoptic area and bed nucleus of the stria terminalis in the rat

    J. Neuroendocrinol.

    (2010)
  • J. Bouchal et al.

    Transcriptional coactivators p300 and CBP stimulate estrogen receptor-beta signaling and regulate cellular events in prostate cancer

    Prostate

    (2011)
  • B. Buranasin

    The effects of rooming-in on the success of breastfeeding and the decline in abandonment of children

    Asia Pac. J. Public Health

    (1991)
  • M.J. Burns et al.

    Standardisation of data from real-time quantitative PCR methods — evaluation of outliers and comparison of calibration curves

    BMC Biotechnol.

    (2005)
  • K. Bystrova et al.

    Early contact versus separation: effects on mother–infant interaction one year later

    Birth

    (2009)
  • G. Calamandrei et al.

    Differential expression of Fos protein in the brain of female mice dependent on pup sensory cues and maternal experience

    Behav. Neurosci.

    (1994)
  • F. Champagne et al.

    Naturally occurring variations in maternal behavior in the rat are associated with differences in estrogen-inducible central oxytocin receptors

    Proc. Natl. Acad. Sci. U. S. A.

    (2001)
  • F.A. Champagne et al.

    Natural variations in maternal care are associated with estrogen receptor alpha expression and estrogen sensitivity in the medial preoptic area

    Endocrinology

    (2003)
  • Cited by (63)

    • Neurobiological mechanisms governing caregiving behavior

      2021, Encyclopedia of Behavioral Neuroscience: Second Edition
    • Changes in maternal motivation across reproductive states in mice: A role for prolactin receptor activation on GABA neurons

      2021, Hormones and Behavior
      Citation Excerpt :

      The University of Otago Animal Ethics Committee approved all animal experimental protocols. The T-maze pup retrieval test was used as a novel environment test for pup-related motivation, as described in the literature (Stolzenberg and Rissman, 2011; Stolzenberg et al., 2012). Four groups of female C57BL/6 J mice were tested in the T-maze pup retrieval test to characterise pup-related motivation behaviour: pup-naïve virgins (n = 6), pregnant day 19 females (n = 8), lactating day 6–7 females (n = 8) and pup-exposed virgins (n = 9).

    View all citing articles on Scopus

    Funding: This work has been supported by the National Institutes of Health T32 DK007646 and R01 MH057759.

    View full text