Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38559030

ABSTRACT

Early-life stress increases sensitivity to subsequent stress, which has been observed among humans, other animals, at the level of cellular activity, and at the level of gene expression. However, the molecular mechanisms underlying such long-lasting sensitivity are poorly understood. We tested the hypothesis that persistent changes in transcription and transcriptional potential were maintained at the level of the epigenome, through changes in chromatin. We used a combination of bottom-up mass spectrometry, viral-mediated epigenome-editing, behavioral quantification, and RNA-sequencing in a mouse model of early-life stress, focusing on the ventral tegmental area (VTA), a brain region critically implicated in motivation, reward learning, stress response, and mood and drug disorders. We find that early-life stress in mice alters histone dynamics in VTA and that a majority of these modifications are associated with an open chromatin state that would predict active, primed, or poised gene expression, including enriched histone-3 lysine-4 methylation and the H3K4 monomethylase Setd7. Mimicking ELS through over-expression of Setd7 and enrichment of H3K4me1 in VTA recapitulates ELS-induced behavioral and transcriptional hypersensitivity to future stress. These findings enrich our understanding of the epigenetic mechanisms linking early-life environmental experiences to long-term alterations in stress reactivity within the brain's reward circuitry, with implications for understanding and potentially treating mood and anxiety disorders in humans.

2.
Horm Behav ; 159: 105472, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38141539

ABSTRACT

Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons, Otx2, shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.


Subject(s)
Adverse Childhood Experiences , Ventral Tegmental Area , Mice , Animals , Male , Female , Ventral Tegmental Area/metabolism , Dopaminergic Neurons/metabolism , Thyroid Hormones/metabolism , Gene Expression , Stress, Psychological/genetics
3.
Proc Natl Acad Sci U S A ; 120(49): e2305776120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011563

ABSTRACT

Individuals with a history of early-life stress (ELS) tend to have an altered course of depression and lower treatment response rates. Research suggests that ELS alters brain development, but the molecular changes in the brain following ELS that may mediate altered antidepressant response have not been systematically studied. Sex and gender also impact the risk of depression and treatment response. Here, we leveraged existing RNA sequencing datasets from 1) blood samples from depressed female- and male-identifying patients treated with escitalopram or desvenlafaxine and assessed for treatment response or failure; 2) the nucleus accumbens (NAc) of female and male mice exposed to ELS and/or adult stress; and 3) the NAc of mice after adult stress, antidepressant treatment with imipramine or ketamine, and assessed for treatment response or failure. We find that transcriptomic signatures of adult stress after a history of ELS correspond with transcriptomic signatures of treatment nonresponse, across species and multiple classes of antidepressants. Transcriptomic correspondence with treatment outcome was stronger among females and weaker among males. We next pharmacologically tested these predictions in our mouse model of early-life and adult social defeat stress and treatment with either chronic escitalopram or acute ketamine. Among female mice, the strongest predictor of behavior was an interaction between ELS and ketamine treatment. Among males, however, early experience and treatment were poor predictors of behavior, mirroring our bioinformatic predictions. These studies provide neurobiological evidence for molecular adaptations in the brain related to sex and ELS that contribute to antidepressant treatment response.


Subject(s)
Adverse Childhood Experiences , Ketamine , Humans , Male , Female , Mice , Animals , Depression/drug therapy , Depression/genetics , Escitalopram , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Treatment Outcome , Stress, Psychological/drug therapy , Stress, Psychological/genetics
4.
bioRxiv ; 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37662236

ABSTRACT

Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.

5.
J Neurosci ; 43(34): 5996-6009, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37429717

ABSTRACT

Early-life stress (ELS) is one of the strongest lifetime risk factors for depression, anxiety, suicide, and other psychiatric disorders, particularly after facing additional stressful events later in life. Human and animal studies demonstrate that ELS sensitizes individuals to subsequent stress. However, the neurobiological basis of such stress sensitization remains largely unexplored. We hypothesized that ELS-induced stress sensitization would be detectable at the level of neuronal ensembles, such that cells activated by ELS would be more reactive to adult stress. To test this, we leveraged transgenic mice to genetically tag, track, and manipulate experience-activated neurons. We found that in both male and female mice, ELS-activated neurons within the nucleus accumbens (NAc), and to a lesser extent the medial prefrontal cortex, were preferentially reactivated by adult stress. To test whether reactivation of ELS-activated ensembles in the NAc contributes to stress hypersensitivity, we expressed hM4Dis receptor in control or ELS-activated neurons of pups and chemogenetically inhibited their activity during experience of adult stress. Inhibition of ELS-activated NAc neurons, but not control-tagged neurons, ameliorated social avoidance behavior following chronic social defeat stress in males. These data provide evidence that ELS-induced stress hypersensitivity is encoded at the level of corticolimbic neuronal ensembles.SIGNIFICANCE STATEMENT Early-life stress enhances sensitivity to stress later in life, yet the mechanisms of such stress sensitization are largely unknown. Here, we show that neuronal ensembles in corticolimbic brain regions remain hypersensitive to stress across the life span, and quieting these ensembles during experience of adult stress rescues stress hypersensitivity.


Subject(s)
Adverse Childhood Experiences , Prefrontal Cortex , Adult , Humans , Male , Mice , Female , Animals , Prefrontal Cortex/physiology , Stress, Psychological/psychology , Neurons , Anxiety , Mice, Transgenic
6.
Horm Behav ; 152: 105364, 2023 06.
Article in English | MEDLINE | ID: mdl-37087766

ABSTRACT

Parental care is diversely demonstrated across the animal kingdom, such that active practitioners and repertoires of parental behavior vary dramatically between and within taxa. For mammals, maternal care is ubiquitous while paternal and alloparental care are rare. The African striped mouse, a rodent species in the family Muridae, demonstrates maternal, paternal, and alloparental care. Because socio-environmental factors can considerably influence the development of their social behavior, including that of paternal and alloparental care, African striped mice are considered socially flexible. Here, we highlight African striped mice as a new model for the neurobiological study of male parental care. We first provide essential background information on the species' natural ecological setting and reproductive behavior, as well as the species-relevant interaction between ecology and reproduction. We then introduce the nature of maternal, paternal, and alloparental care in the species. Lastly, we provide a review of existing developmental and neurobiological perspectives and highlight potential avenues for future research.


Subject(s)
Murinae , Social Behavior , Animals , Mice , Male , Humans , Reproduction , Fathers
7.
Dev Psychobiol ; 64(1): e22227, 2022 01.
Article in English | MEDLINE | ID: mdl-35050507

ABSTRACT

We explored the associations between early-life adversity and migration-related stress on the mental health of Central American and Mexican migrating children held in United States immigration detention facilities. Migrating children have high rates of trauma exposure prior to and during migration. Early-life adversity increases risk for developing mental health disorders. Forced separation of migrating children from their parents at the United States-Mexico border potentially exacerbates this risk. We sought to determine whether exposure to trauma prior to immigration and specific features of immigration detention were associated with posttraumatic stress symptomatology. We interviewed parents of 84 migrating children (ages 1-17) after families were released from immigration detention facilities to assess children's migration- and detention-related experiences. A modified version of the University of California Los Angeles Posttraumatic Stress Disorder (PTSD) Reaction Index was administered to assess children's PTSD symptoms and document trauma exposure. A total of 97.4% of children experienced at least one premigration traumatic event. PTSD symptom severity was most strongly predicted by premigration trauma and duration of parent-child separation. This study contributes to a growing empirical literature documenting that early-life adversity increases risk of developing mental health disorders, particularly following additional stress exposure, and that remaining with parents during immigration detention may help mitigate children's stress response.


Subject(s)
Emigration and Immigration , Hispanic or Latino , Adolescent , Central America , Child , Child, Preschool , Humans , Infant , Mexico , Outcome Assessment, Health Care , United States
8.
Biol Psychiatry ; 91(1): 36-42, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33602500

ABSTRACT

Both history of early-life stress (ELS) and female sex are associated with increased risk for depression. The complexity of how ELS interacts with brain development and sex to impart risk for multifaceted neuropsychiatric disorders is also unlikely to be understood by examining changes in single genes. Here, we review an emerging literature on genome-wide transcriptional and epigenetic signatures of ELS and the potential moderating influence of sex. We discuss evidence both that there are latent sex differences revealed by ELS and that ELS itself produces latent transcriptomic changes revealed by adult stress. In instances where there are broad similarities in global signatures of ELS among females and males, genes that contribute to these patterns are largely distinct based on sex. As this area of investigation grows, an effort should be made to better understand the sex-specific impact of ELS within the human brain, specific contributions of chromosomal versus hormonal sex, how ELS alters the time course of normal transcriptional development, and the cell-type specificity of transcriptomic and epigenomic changes in the brain. A better understanding of how ELS interacts with sex to alter transcriptomic and epigenomic signatures in the brain will inform individualized therapeutic strategies to prevent or ameliorate depression and other psychiatric disorders in this vulnerable population.


Subject(s)
Adverse Childhood Experiences , Brain , Epigenomics , Female , Humans , Male , Sex Characteristics , Stress, Psychological/genetics
9.
Nat Commun ; 10(1): 5098, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704941

ABSTRACT

Abuse, neglect, and other forms of early life stress (ELS) significantly increase risk for psychiatric disorders including depression. In this study, we show that ELS in a postnatal sensitive period increases sensitivity to adult stress in female mice, consistent with our earlier findings in male mice. We used RNA-sequencing in the ventral tegmental area, nucleus accumbens, and prefrontal cortex of male and female mice to show that adult stress is distinctly represented in the brain's transcriptome depending on ELS history. We identify: 1) biological pathways disrupted after ELS and associated with increased behavioral stress sensitivity, 2) putative transcriptional regulators of the effect of ELS on adult stress response, and 3) subsets of primed genes specifically associated with latent behavioral changes. We also provide transcriptomic evidence that ELS increases sensitivity to future stress through enhancement of known programs of cortical plasticity.


Subject(s)
Maternal Deprivation , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Reward , Stress, Psychological/genetics , Transcriptome , Ventral Tegmental Area/metabolism , Animals , Depression/genetics , Female , Gene Expression Profiling , Housing, Animal , Male , Mice , Sequence Analysis, RNA
10.
Front Behav Neurosci ; 13: 40, 2019.
Article in English | MEDLINE | ID: mdl-30881296

ABSTRACT

Epidemiological evidence identifies early life adversity as a significant risk factor for the development of mood disorders. Much evidence points to the role of early life experience in susceptibility and, to a lesser extent, resilience, to stress in adulthood. While many models of these phenomena exist in the literature, results are often conflicting and a systematic comparison of multiple models is lacking. Here, we compare effects of nine manipulations spanning the early postnatal through peri-adolescent periods, both at baseline and following exposure to chronic social defeat stress in adulthood, in male mice. By applying rigorous criteria across three commonly used measures of depression- and anxiety-like behavior, we identify manipulations that increase susceptibility to subsequent stress in adulthood and other pro-resilient manipulations that mitigate the deleterious consequences of adult stress. Our findings point to the importance of timing of early life stress and provide the foundation for future studies to probe the neurobiological mechanisms of risk and resilience conferred by variation in the early life environment.

12.
Nat Neurosci ; 18(3): 415-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25643298

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has a crucial role in modulating neural and behavioral plasticity to drugs of abuse. We found a persistent downregulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which was mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increased stalling of RNA polymerase II at these Bdnf promoters in VTA and altered permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we found that morphine suppressed binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which resulted from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributed to Bdnf repression and associated behavioral plasticity to morphine. Our findings suggest previously unknown epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.


Subject(s)
Analgesics, Opioid/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Epigenesis, Genetic/physiology , Ventral Tegmental Area/metabolism , Analgesics, Opioid/pharmacology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cocaine/pharmacology , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic/drug effects , Heroin Dependence/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Postmortem Changes , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Rats , Rats, Sprague-Dawley , Ventral Tegmental Area/drug effects
13.
Dev Neurobiol ; 75(10): 1114-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25044746

ABSTRACT

Maternal behavior is dependent on estrogen receptor-alpha (ERα; Esr1) and oxytocin receptor (OTR) signaling in the medial preoptic area (MPOA) of the hypothalamus, as well as dopamine signaling from the ventral tegmental area (VTA) to forebrain regions. Previous studies in rats indicate that low levels of maternal care, particularly licking/grooming (LG), lead to reduced levels of MPOA ERα and VTA dopamine neurons in female offspring and predict lower levels of postpartum maternal behavior by these offspring. The aim of this study was to determine the functional impact on maternal behavior of neonatal manipulation of ERα in females that had experienced low versus high levels of postnatal maternal LG. Adenovirus expressing ESR1 was targeted to the MPOA in female pups from low and high LG litters on postnatal day 2-3. Overexpression of ESR1 in low LG offspring elevated the level of ERα-immunoreactive cells in the MPOA and of tyrosine hydroxylase cells in the VTA to that observed in high LG females. Amongst juvenile female low LG offspring, ESR1 overexpression also decreased the latency to engage in maternal behavior toward donor pups. These results show that virally mediated expression of ESR1 in the neonatal rat hypothalamus results in lasting changes in ESR1 expression through the juvenile period, and can "rescue" hormone receptor levels and behavior of offspring reared by low LG dams, potentially mediated by downstream alterations within reward circuitry. Thus, the transmission of maternal behavior from one generation to the next can be augmented by neonatal ERα in the MPOA.


Subject(s)
Behavior, Animal/physiology , Dopaminergic Neurons/metabolism , Estrogen Receptor alpha/metabolism , Maternal Behavior/physiology , Mesencephalon/metabolism , Animals , Animals, Newborn , Dopamine/metabolism , Female , Grooming/physiology , Hypothalamus/growth & development , Hypothalamus/metabolism , Rats, Long-Evans , Reward , Ventral Tegmental Area/growth & development , Ventral Tegmental Area/metabolism
14.
Neuropsychopharmacology ; 39(11): 2646-53, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24853771

ABSTRACT

The nucleus accumbens (NAc) has a central role in the mechanism of action of drugs of abuse. The major neuronal type within the NAc is the GABAergic medium spiny neuron (MSN), with two major subpopulations defined-termed D1-type and D2-type MSNs-based on the predominant dopamine receptor expressed. However, very little is known about the contribution of altered GABAergic function in NAc MSNs to the neural and behavioral plasticity that contributes to the lasting actions of drugs of abuse. In the present study, we show that GABAergic activity is selectively modulated in D1-type MSNs of the NAc by signaling of brain-derived neurotrophic factor (BDNF) and its receptor, tyrosine receptor kinase B (TrkB), and that such adaptations control rewarding responses to morphine. Optical activation of D1-type MSNs, or the knockout of TrkB from D1-type MSNs (D1-TrkB KO), enhances morphine reward, effects not seen for D2-type MSNs. In addition, D1-TrkB KO mice, but not D2-TrkB KO mice, display decreased GABAA receptor (GABAAR) subunit expression and reduced spontaneous inhibitory postsynaptic currents (sIPSCs) in D1-type, but not D2-type, MSNs in the NAc. Furthermore, we found that GABAAR antagonism in the NAc enhances morphine reward and that morphine exposure decreases TrkB expression as well as GABAergic activity in D1-type MSNs. Together, these data provide evidence for the enhancement of morphine reward through reduction of inhibitory GABAAR responses, an adaptation mediated by morphine-induced reduction of BDNF-TrkB signaling in D1-type MSNs.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , GABAergic Neurons/drug effects , Morphine/pharmacology , Narcotics/pharmacology , Nucleus Accumbens/drug effects , Receptors, Dopamine D1/metabolism , Animals , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/physiology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Mice, Inbred C57BL , Mice, Knockout , Nucleus Accumbens/physiology , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, Dopamine D2/metabolism , Receptors, GABA-A/metabolism , Reward , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
15.
Eur J Neurosci ; 39(6): 946-956, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24446918

ABSTRACT

Variation within mesolimbic dopamine (DA) pathways has significant implications for behavioral responses to rewards, and previous studies have indicated long-term programming effects of early life stress on these pathways. In the current study, we examined the impact of natural variations in maternal care in Long Evans rats on the development of DA pathways in female offspring and the consequences for reward-directed behaviors. We found that tyrosine hydroxylase (TH) immunoreactivity in the ventral tegmental area was elevated by postnatal day 6 in response to maternal licking/grooming (LG), and that these effects were sustained into adulthood. Increased TH immunoreactivity was not found to be associated with altered epigenetic regulation or transcriptional activation of Th, but probably involved LG-associated changes in the differentiation of postnatal DA neurons through increased expression of Cdkn1c, and enhanced survival of DA projections through LG-associated increases in Lmx1b and brain-derived neurotrophic factor. At weaning, high-LG offspring had elevated DA receptor mRNA levels within the nucleus accumbens and increased conditioned place preference for a high-fat diet. In contrast, high-LG, as compared with low-LG, juvenile offspring had a reduced preference for social interactions with siblings, and haloperidol administration abolished group differences in conditioned place preference through a shift towards increased social preferences in high-LG offspring. The effects of maternal care on developing DA pathways and reward-directed behavior of female offspring that we have observed may play a critical role in the behavioral transmission of maternal LG from mother to daughter, and account for individual differences in the mesolimbic DA system.


Subject(s)
Dopaminergic Neurons/metabolism , Maternal Behavior , Mesencephalon/growth & development , Reward , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mesencephalon/metabolism , Mesencephalon/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
16.
Endocrinology ; 154(11): 4340-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002038

ABSTRACT

Maternal care experienced during postnatal development has enduring effects on neuroendocrine function and behavior. Previous studies in rats have illustrated the effect of maternal licking/grooming (LG) on hormone receptors and maternal behavior of adult female offspring associated with altered DNA methylation. However, the developmental timing of these effects, which provide insight into the cellular and molecular pathways through which early experience alters later behavior, had not been explored. Here, we demonstrate the developmental emergence of these outcomes and use cross-fostering to identify sensitive periods for these effects. Estrogen receptor (ER)α and ERß mRNA levels within the medial preoptic area (MPOA) of the hypothalamus were increased by postnatal day (PN)21 in female offspring of high LG dams; LG-associated increases in oxytocin receptor mRNA levels were observed beyond the weaning period. Quantification of ERα-immunoreactivity indicated a high degree of neuroanatomical specificity of LG effects within the MPOA that were observed by PN6. Reduced DNA methylation and histone 3 lysine 9 tri-methylation and increased histone 3 lysine 4 tri-methylation at the ERα gene promoter (Esr1) were detected at PN21 in high LG female offspring. Latency to engage in maternal behavior toward donor pups was significantly shorter among high LG females. Cross-fostering revealed that maternal sensitization and MPOA ERα levels are sensitive to maternal care experienced before but not after PN10. Differential windows of plasticity were identified for ERß and oxytocin receptor mRNA levels. These studies contribute significantly to our understanding of the molecular, neurobiological, and behavioral pathways through which variation in maternal behavior is transmitted from one generation to the next.


Subject(s)
Behavior, Animal/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Maternal Behavior/physiology , Receptors, Steroid/metabolism , Animals , DNA Methylation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Hypothalamus , Rats , Rats, Inbred LEC , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Receptors, Steroid/genetics , Sexual Maturation
SELECTION OF CITATIONS
SEARCH DETAIL
...