Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Behav Brain Sci ; 46: e216, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37695011

RESUMO

We generally agree with Burt's thesis. However, we note that the author did not discuss epigenetics, the study of how the environment can alter gene structure and function. Given epigenetic mechanisms, the utility of polygenic risk scores (PRS) is limited in studies of development and mental illness. Finally, in this commentary we expand upon the risks of reliance upon PRSs.


Assuntos
Transtornos Mentais , Psiquiatria , Humanos , Epigênese Genética , Transtornos Mentais/genética , Fatores de Risco
2.
Horm Behav ; 121: 104709, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32007517

RESUMO

Territoriality is an adaptive behavioral trait that is important for animal's fitness and there still remains much to learn about the proximate mechanisms underlying the development of territoriality. We speculate that the formation of a conditioned place preference (CPP), an increased time allocation to the environment where a rewarding experience occurred, contributes to territoriality. Testosterone (T) plays an important role in modulating territorial behaviors and T pulses can induce a CPP. We confirmed previous findings in California mice (Peromyscus californicus) that T pulses can induce a CPP in singly-housed, but not group-housed males. Housing singly may be similar enough to dispersal in nature to initiate similar hormonal and neuroanatomical changes needed for the development of territoriality. We further revealed that T pulses interact with the single housing experience and appear to enhance the motivation to be aggressive towards a stimulus male. On a neural level, being singly housed upregulated levels of androgen receptors in the preoptic area, which positively correlated with the strength of the CPP. We speculate that this change in androgen sensitivity in the preoptic area is characteristic of males that have dispersed, making them more sensitive to T pulses. Also, single housing increased markers of synaptic plasticity in the nucleus accumbens, ventral and dorsal hippocampus, neural changes that may be associated with dispersal, reproduction and territory establishment. These behavioral and neural changes may reflect the life history transition from residing in the natal territory to dispersing and establishing a new territory.


Assuntos
Comportamento Animal/efeitos dos fármacos , Modelos Biológicos , Peromyscus/fisiologia , Territorialidade , Testosterona/farmacologia , Agressão/efeitos dos fármacos , Animais , Comportamento de Escolha/efeitos dos fármacos , Condicionamento Psicológico/efeitos dos fármacos , Abrigo para Animais , Masculino , Motivação/efeitos dos fármacos , Comportamento de Nidação/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Receptores Androgênicos/metabolismo , Reprodução/efeitos dos fármacos , Recompensa , Testosterona/fisiologia
3.
J Pediatr ; 172: 20-8, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26970931

RESUMO

OBJECTIVE: To determine the impact of maternal obesity and gestational weight gain across pregnancy on fetal indices of inflammation and iron status. STUDY DESIGN: Eighty-five healthy term newborns delivered via elective cesarean were categorized by 2 maternal body mass index (BMI) thresholds; above or below 30 kg/m(2) or above or below 35 kg/m(2). Umbilical cord plasma levels of C-reactive protein, interleukin (IL)-6, tumor necrosis factor (TNF)-α, ferritin, and hepcidin were assayed. Cytokines released by phytohemagglutinin-stimulated umbilical cord mononuclear cells (MNCs) were assayed. RESULTS: Maternal class II obesity, defined as BMI of 35 kg/m(2) and above, predicted higher C-reactive protein and TNF-α in umbilical cord plasma (P < .05 for both), and also proinflammatory cytokines (IL-1ß, IL-6, and TNF-α) from stimulated MNC (P < .05 for all). The rise in plasma TNF-α and MNC TNF-α was not linear but occurred when the threshold of BMI 35 kg/m(2) was reached (P < .005, P < .06). Poorer umbilical cord iron indices were associated with maternal obesity. When ferritin was low, IL-6 was higher (P < .04), but this relationship was present primarily when maternal BMI exceeded 35 kg/m(2) (P < .03). Ferritin was correlated with hepcidin (P < .0001), but hepcidin was unrelated to either maternal BMI or inflammatory indices. CONCLUSIONS: Class II obesity and above during pregnancy is associated with fetal inflammation in a threshold fashion. Although maternal BMI negatively impacted fetal iron status, hepcidin, related to obesity in adults, was related to iron status and not obesity in fetuses. Pediatricians should be aware of these relationships.


Assuntos
Citocinas/sangue , Sangue Fetal/metabolismo , Inflamação/metabolismo , Ferro/sangue , Obesidade/sangue , Adulto , Índice de Massa Corporal , Feminino , Feto/metabolismo , Humanos , Recém-Nascido , Inflamação/complicações , Masculino , Troca Materno-Fetal , Gravidez
4.
Brain Behav Immun ; 46: 60-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25728234

RESUMO

Precise regulation of the epigenome during perinatal development is critical to the formation of species-typical behavior later in life. Recent data suggests that Gadd45b facilitates active DNA demethylation by recruiting proteins involved in base excision repair (BER), which will catalyze substitution of 5-methyl-cytosine (5mC) for an unmodified cytosine. While a role for Gadd45b has been implicated in both hippocampal and amygdalar learning tasks, to the best of our knowledge, no study has been done investigating the involvement of Gadd45b in neurodevelopmental programming of social behavior. To address this, we used a targeted siRNA delivery approach to transiently knock down Gadd45b expression in the neonatal rat amygdala. We chose to examine social behavior in the juvenile period, as social deficits associated with neurodevelopmental disorders tend to emerge in humans at an equivalent age. We find that neonatal Gadd45b knock-down results in altered juvenile social behavior and reduced expression of several genes implicated in psychiatric disorders, including methyl-CpG-binding protein 2 (MeCP2), Reelin, and brain derived neurotrophic factor (BDNF). We furthermore report a novel role for Gadd45b in the programmed expression of α2-adrenoceptor (Adra2a). Consistent with Gadd45b's role in the periphery, we also observed changes in the expression of pro-inflammatory cytokines interleukin-6 (Il-6) and interleukin-1beta (Il-1beta) in the amygdala, which could potentially mediate or exacerbate effects of Gadd45b knockdown on the organization of social behavior. These data suggest a prominent role for Gadd45b in the epigenetic programming of complex juvenile social interactions, and may provide insight into the etiology of juvenile behavioral disorders such as ADHD, autism, and/or schizophrenia.


Assuntos
Tonsila do Cerebelo/metabolismo , Antígenos de Diferenciação/metabolismo , Comportamento Animal/fisiologia , Citocinas/metabolismo , Epigênese Genética , Comportamento Social , Animais , Antígenos de Diferenciação/genética , Ansiedade/genética , Ansiedade/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Inflamação/genética , Inflamação/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Proteína Reelina , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
5.
Front Neuroendocrinol ; 34(3): 190-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23707698

RESUMO

The emerging area of neuroepigenetics has been linked to numerous mental health illnesses. Importantly, a large portion of what we know about early gene×environment interactions comes from examining epigenetic modifications of neuroendocrine systems. This review will highlight how neuroepigenetic mechanisms during brain development program lasting differences in neuroendocrine systems and how other neuroepigenetic processes remain plastic, even within the adult brain. As epigenetic mechanisms can either be stable or plastic, elucidating the mechanisms involved in reversing these processes could aid in understanding how to reverse pathological epigenetic programming.


Assuntos
Epigênese Genética , Plasticidade Neuronal/fisiologia , Diferenciação Sexual/genética , Encéfalo/crescimento & desenvolvimento , Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Epigenômica , Histonas/metabolismo , Neuroendocrinologia
6.
Proc Natl Acad Sci U S A ; 108(10): 4242-7, 2011 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-21368111

RESUMO

Although some DNA methylation patterns are altered by steroid hormone exposure in the developing brain, less is known about how changes in steroid hormone levels influence DNA methylation patterns in the adult brain. Steroid hormones act in the adult brain to regulate gene expression. Specifically, the expression of the socially relevant peptide vasopressin (AVP) within the bed nucleus of the stria terminalis (BST) of adult brain is dependent upon testosterone exposure. Castration dramatically reduces and testosterone replacement restores AVP expression within the BST. As decreases in mRNA expression are associated with increases in DNA promoter methylation, we explored the hypothesis that AVP expression in the adult brain is maintained through sustained epigenetic modifications of the AVP gene promoter. We find that castration of adult male rats resulted in decreased AVP mRNA expression and increased methylation of specific CpG sites within the AVP promoter in the BST. Similarly, castration significantly increased estrogen receptor α (ERα) mRNA expression and decreased ERα promoter methylation within the BST. These changes were prevented by testosterone replacement. This suggests that the DNA promoter methylation status of some steroid responsive genes in the adult brain is actively maintained by the presence of circulating steroid hormones. The maintenance of methylated or demethylated states of some genes in the adult brain by the presence of steroid hormones may play a role in the homeostatic regulation of behaviorally relevant systems.


Assuntos
Arginina Vasopressina/metabolismo , Encéfalo/metabolismo , Epigênese Genética , Hormônios/metabolismo , Esteroides/metabolismo , Animais , Arginina Vasopressina/genética , Sequência de Bases , DNA , Metilação de DNA , Receptor alfa de Estrogênio/genética , Masculino , Dados de Sequência Molecular , Orquiectomia , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Ratos
7.
Front Mol Neurosci ; 17: 1322273, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38486962

RESUMO

The use of predators and predator odor as stressors is an important and ecologically relevant model for studying the impact of behavioral responses to threat. Here we summarize neural substrates and behavioral changes in rats resulting from predator exposure. We briefly define the impact predator exposure has on neural targets throughout development (neonatal, juvenile, and adulthood). These findings allow us to conceptualize the impact of predator exposure in the brain, which in turn may have broader implications for human disorders such as PTSD. Importantly, inclusion of sex as a biological variable yields distinct results that may indicate neural substrates impacted by predator exposure differ based on sex.

8.
Endocrinology ; 165(5)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38597659

RESUMO

We examined the effect of the puberty blocker, leuprolide acetate, on sex differences in juvenile rough-and-tumble play behavior and anxiety-like behavior in adolescent male and female rats. We also evaluated leuprolide treatment on gonadal and pituitary hormone levels and activity-regulated cytoskeleton-protein messenger RNA levels within the adolescent amygdala, a region important both for rough-and-tumble play and anxiety-like behavior. Our findings suggest that leuprolide treatment lowered anxiety-like behavior during adolescent development, suggesting that the maturation of gonadotropin-releasing hormone systems may be linked to increased anxiety. These data provide a potential new model to understand the emergence of increased anxiety triggered around puberty. Leuprolide also reduced masculinized levels of rough-and-tumble play behavior, lowered follicle-stimulating hormone, and produced a consistent pattern of reducing or halting sex differences of hormone levels, including testosterone, growth hormone, thyrotropin, and corticosterone levels. Therefore, leuprolide treatment not only pauses sexual development of peripheral tissues, but also reduces sex differences in hormones, brain, and behavior, allowing for better harmonization of these systems following gender-affirming hormone treatment. These data contribute to the intended use of puberty blockers in stopping sex differences from developing further with the potential benefit of lowering anxiety-like behavior.


Assuntos
Ansiedade , Comportamento Animal , Leuprolida , Maturidade Sexual , Animais , Leuprolida/farmacologia , Masculino , Feminino , Ansiedade/tratamento farmacológico , Ratos , Comportamento Animal/efeitos dos fármacos , Maturidade Sexual/efeitos dos fármacos , Caracteres Sexuais , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Corticosterona/sangue , Ratos Sprague-Dawley , Testosterona/sangue
9.
Pflugers Arch ; 465(5): 573-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23503727

RESUMO

The concept that the brain differs in make-up between males and females is not new. For example, it is well established that anatomists in the nineteenth century found sex differences in human brain weight. The importance of sex differences in the organization of the brain cannot be overstated as they may directly affect cognitive functions, such as verbal skills and visuospatial tasks in a sex-dependent fashion. Moreover, the incidence of neurological and psychiatric diseases is also highly dependent on sex. These clinical observations reiterate the importance that gender must be taken into account as a relevant possible contributing factor in order to understand the pathogenesis of neurological and psychiatric disorders. Gender-dependent differentiation of the brain has been detected at every level of organization--morphological, neurochemical, and functional--and has been shown to be primarily controlled by sex differences in gonadal steroid hormone levels during perinatal development. In this review, we discuss howthe gonadal steroid hormone testosterone and its metabolites affect downstream signaling cascades, including gonadal steroid receptor activation, and epigenetic events in order to differentiate the brain in a gender-dependent fashion.


Assuntos
Encéfalo/crescimento & desenvolvimento , Epigênese Genética , Caracteres Sexuais , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Feminino , Hormônios Esteroides Gonadais/metabolismo , Humanos , Masculino
10.
Proc Natl Acad Sci U S A ; 107(27): 12393-8, 2010 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-20616093

RESUMO

Winning aggressive disputes can enhance future fighting ability and the desire to seek out additional contests. In some instances, these effects are long lasting and vary in response to the physical location of a fight. Thus, in principle, winning aggressive encounters may cause long-term and context-dependent changes to brain areas that control the output of antagonistic behavior or the motivation to fight (or both). We examined this issue in the territorial California mouse (Peromyscus californicus) because males of this species are more likely to win fights after accruing victories in their home territory but not after accruing victories in unfamiliar locations. Using immunocytochemistry and real-time quantitative PCR, we found that winning fights either at home or away increases the expression of androgen receptors (AR) in the medial anterior bed nucleus of the stria terminalis, a key brain area that controls social aggression. We also found that AR expression in brain regions that mediate motivation and reward, nucleus accumbens (NAcc) and ventral tegmental area (VTA), increases only in response to fights in the home territory. These effects of winning were likely exclusive to the neural androgenic system because they have no detectible impact on the expression of progestin receptors. Finally, we demonstrated that the observed changes in androgen sensitivity in the NAcc and VTA are positively associated with the ability to win aggressive contests. Thus, winning fights can change brain phenotype in a manner that likely promotes future victory and possibly primes neural circuits that motivate individuals to fight.


Assuntos
Agressão/fisiologia , Motivação/fisiologia , Vias Neurais/fisiologia , Receptores Androgênicos/fisiologia , Animais , Comportamento Competitivo/fisiologia , Dominação-Subordinação , Feminino , Imuno-Histoquímica , Masculino , Peromyscus , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Núcleos Septais/metabolismo , Comportamento Social , Meio Social , Territorialidade
11.
Neuropsychopharmacology ; 48(13): 1952-1962, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37640922

RESUMO

Mu-opioid receptor (µ-OR) signaling in forebrain sites including nucleus accumbens (Acb) and ventromedial prefrontal cortex (vmPFC) modulates reward-driven feeding and may play a role in the pathophysiology of disordered eating. In preclinical models, intra-Acb or intra-vmPFC µ-OR stimulation causes overeating and vigorous responding for food rewards. These effects have been studied mainly in male animals, despite demonstrated sex differences and estrogen modulation of central reward systems. Hence, the present study investigated sex differences and estrogen modulation of intra-Acb and intra-vmPFC µ-OR-driven feeding behaviors. First, the dose-related effects of intra-Acb and intra-vmPFC infusions of the µ-OR-selective agonist, DAMGO, were compared among intact female, ovariectomized (OVX) female, and intact male rats. The DAMGO feeding dose-effect function was flattened in intact females relative to the robust, dose-dependent effects observed in OVX females and intact males. Thus, in intact females, intra-Acb DAMGO failed to elevate food intake relative to vehicle, while intra-vmPFC DAMGO elevated food intake, but to a smaller degree compared to males and OVX females. Next, to explore the possible role of estrogen in mediating the diminished DAMGO response observed in intact females, OVX rats were given intra-Acb or intra-vmPFC infusions of DAMGO either immediately after a subcutaneous injection of 17-beta-estradiol 3-benzoate (EB; 5 µg/0.1 mL) or 24 h after EB injection. Intra-Acb DAMGO effects were not changed at the immediate post-EB time point. At the delayed post-EB timepoint, significant lordosis was noted and the duration of intra-Acb DAMGO-driven feeding bouts was significantly reduced, with no change in the number of bouts initiated, locomotor hyperactivity, or Fos immunoreactivity in hypothalamic feeding and arousal systems. Similarly, EB failed to alter the motor-activational effects of intra-vmPFC DAMGO while reducing feeding. These findings indicate that delayed, presumably genomically mediated estrogen actions modulate the µ-OR-generated motivational state by reducing consummatory activity while sparing goal-approach and general arousal/activity. The results additionally suggest that EB regulation of consummatory activity occurs outside of forebrain-µ-OR control of hypothalamic systems.


Assuntos
Analgésicos Opioides , Comportamento Alimentar , Ratos , Feminino , Masculino , Animais , Analgésicos Opioides/farmacologia , Ratos Sprague-Dawley , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Núcleo Accumbens , Estrogênios/farmacologia , Atividade Motora , Receptores Opioides mu/metabolismo
12.
Brain Res ; 1809: 148339, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36966960

RESUMO

DNA topoisomerases are essential for preserving genomic integrity. DNA topoisomerases induce breakage of DNA to facilitate replication and transcription by relaxing DNA and relieving supercoiling. Aberrant expression and deletions of topoisomerases are associated with psychiatric disorders such as schizophrenia and autism. Our study investigated the effects of early life stress (ELS) on three topoisomerases, Top1, Top3α, and Top3ß in the developing rat brain. Newborn rats were exposed to a predator odor stress on postnatal days 1, 2, and 3; brain tissue was collected either 30 min after the last stressor on postnatal day 3 or during the juvenile period. We found that exposure to predator odor resulted in a decrease in Top3ß expression levels in the neonatal male amygdala and in the juvenile prefrontal cortex of males and females. These data suggest that developing males and females respond differently to predator odor-induced stress. As ELS results in lower Top3ß levels, these data suggest that ELS experienced during development may have consequences for genomic structural integrity and increased mental health risk.


Assuntos
DNA Topoisomerases Tipo I , Estresse Psicológico , Animais , Feminino , Masculino , Ratos , Encéfalo/metabolismo , DNA , DNA Topoisomerases/metabolismo , DNA Topoisomerases Tipo I/química , DNA Topoisomerases Tipo I/genética , DNA Topoisomerases Tipo I/metabolismo , Estresse Psicológico/metabolismo
13.
Brain Behav Immun ; 25(7): 1299-304, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21352906

RESUMO

Variations in maternal care alter the developmental programming of some genes by creating lasting differences in DNA methylation patterns, such as the estrogen receptor alpha (ERα) promoter region. Interestingly, mother rats preferentially lick and groom their male offspring more than females; therefore, we questioned whether the somatosensory stimuli associated with maternal grooming influences potential sex differences in DNA methylation patterns within the developing amygdala, an area important for socioemotional processing. We report a sex difference in the DNA methylation pattern of specific CpG sites of the ERα promoter region within the developing amygdala. Specifically, males have higher levels of ERα promoter methylation contrasted to females. Increasing the levels of maternal stimuli in females masculinized ERα promoter methylation patterns to male-like levels. As expected, higher levels of ERα promoter methylation were associated with lower ERα mRNA levels. These data provide further evidence that the early neonatal environment, particularly maternal care, contributes to sex differences and early programming of the neonatal brain via an epigenetic mechanism.


Assuntos
Tonsila do Cerebelo/metabolismo , Epigênese Genética , Receptor alfa de Estrogênio/metabolismo , Asseio Animal , Comportamento Materno/fisiologia , Caracteres Sexuais , Tonsila do Cerebelo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Metilação de DNA , Feminino , Masculino , Regiões Promotoras Genéticas , Ratos , Ratos Sprague-Dawley
14.
Horm Behav ; 59(3): 358-63, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20619265

RESUMO

The study of epigenetic mechanisms is important for elucidating how gene-by-environment interactions can have lasting outcomes on brain function and behavior. In general, studies of epigenetic processes mainly focus on the methylation status of DNA. While methylation of DNA alone can interfere with gene transcription, it is the binding of methyl-CpG binding proteins to methylated DNA, and subsequent recruitment of nuclear corepressors and histone deacetylases, that results in more efficient gene repression. In this review, we will discuss sex differences in DNA methylation patterns, methyl binding proteins, and corepressor proteins that contribute to lasting differences in brain and juvenile behavior. Specifically, we will discuss new data on sex differences in ERα DNA promoter methylation patterns, and the role of MeCP2 and the nuclear corepressor, NCoR, on the organization of juvenile social play behavior.


Assuntos
Comportamento Animal/fisiologia , Epigênese Genética , Jogos e Brinquedos , Caracteres Sexuais , Comportamento Social , Animais , Metilação de DNA , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo
15.
Sci Rep ; 11(1): 3549, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33574362

RESUMO

Early life stress (ELS) has been shown to have a significant impact on typical brain development and the manifestation of psychological disorders through epigenetic modifications that alter gene expression. Line1, a retrotransposon associated with genetic diversity, has been linked with various psychological disorders that are associated with ELS. Our previous work demonstrated altered Line1 DNA copy number in the neonatal period following stressful experiences; we therefore chose to investigate whether early life stress altered Line1 retrotransposition persists into the juvenile period of development. Our study uses a neonatal predator odor exposure (POE) paradigm to model ELS in rats. We examined Line1 using qPCR to assess Line1 expression levels and DNA copy number in the male and female juvenile amygdala, hippocampus and prefrontal cortex-areas chosen for their association with affective disorders and stress. We report a sex difference in Line1 levels within the juvenile amygdala. We also find that ELS significantly increases Line1 DNA copy number within the juvenile amygdala which correlates with reduced juvenile social play levels, suggesting the possibility that Line1 may influence juvenile social development.


Assuntos
Experiências Adversas da Infância , Encéfalo/crescimento & desenvolvimento , Elementos Nucleotídeos Longos e Dispersos/genética , Estresse Psicológico/genética , Tonsila do Cerebelo/crescimento & desenvolvimento , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Animais , Encéfalo/patologia , Variações do Número de Cópias de DNA/genética , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Masculino , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Ratos , Caracteres Sexuais , Estresse Psicológico/patologia
16.
J Neurosci ; 29(41): 12815-23, 2009 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-19828794

RESUMO

Epigenetic changes in the nervous system are emerging as a critical component of enduring effects induced by early life experience, hormonal exposure, trauma and injury, or learning and memory. Sex differences in the brain are largely determined by steroid hormone exposure during a perinatal sensitive period that alters subsequent hormonal and nonhormonal responses throughout the lifespan. Steroid receptors are members of a nuclear receptor transcription factor superfamily and recruit multiple proteins that possess enzymatic activity relevant to epigenetic changes such as acetylation and methylation. Thus steroid hormones are uniquely poised to exert epigenetic effects on the developing nervous system to dictate adult sex differences in brain and behavior. Sex differences in the methylation pattern in the promoter of estrogen and progesterone receptor genes are evident in newborns and persist in adults but with a different pattern. Changes in response to injury and in methyl-binding proteins and steroid receptor coregulatory proteins are also reported. Many steroid-induced epigenetic changes are opportunistic and restricted to a single lifespan, but new evidence suggests endocrine-disrupting compounds can exert multigenerational effects. Similarly, maternal diet also induces transgenerational effects, but the impact is sex specific. The study of epigenetics of sex differences is in its earliest stages, with needed advances in understanding of the hormonal regulation of enzymes controlling acetylation and methylation, coregulatory proteins, transient versus stable DNA methylation patterns, and sex differences across the epigenome to fully understand sex differences in brain and behavior.


Assuntos
Encéfalo/fisiologia , Epigênese Genética/fisiologia , Caracteres Sexuais , Animais , Metilases de Modificação do DNA/metabolismo , Feminino , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Modelos Biológicos
17.
Front Neuroendocrinol ; 30(3): 328-42, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19401208

RESUMO

Steroid hormones act in brain and throughout the body to regulate a variety of functions, including development, reproduction, stress and behavior. Many of these effects of steroid hormones are mediated by their respective receptors, which are members of the steroid/nuclear receptor superfamily of transcriptional activators. A variety of studies in cell lines reveal that nuclear receptor coregulators are critical in modulating steroid receptor-dependent transcription. Thus, in addition to the availability of the hormone and the expression of its receptor, nuclear receptor coregulators are essential for efficient steroid-dependent transactivation of genes. This review will highlight the importance of nuclear receptor coregulators in modulating steroid-dependent gene expression in brain and the regulation of behavior.


Assuntos
Comportamento/fisiologia , Encéfalo/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Transativadores/fisiologia , Animais , Encéfalo/fisiologia , Humanos , Modelos Biológicos , Receptores Citoplasmáticos e Nucleares/metabolismo , Ativação Transcricional/fisiologia
18.
Genes Brain Behav ; 19(7): e12662, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32388931

RESUMO

Neural systems underlying important behaviors are usually highly conserved across species. The medial preoptic area (MPOA) has been demonstrated to play a crucial role in reward associated with affiliative, nonsexual, social communication in songbirds. However, the role of MPOA in affiliative, rewarding social behaviors (eg, social play behavior) in mammals remains largely unknown. Here we applied our insights from songbirds to rats to determine whether opioids in the MPOA govern social play behavior in rats. Using an immediate early gene (ie, Egr1, early growth response 1) expression approach, we identified increased numbers of Egr1-labeled cells in the MPOA after social play in adolescent male rats. We also demonstrated that cells expressing mu opioid receptors (MORs, gene name Oprm1) in the MPOA displayed increased Egr1 expression when adolescent rats were engaged in social play using double immunofluorescence labeling of MOR and Egr1. Furthermore, using short hairpin RNA-mediated gene silencing we revealed that knockdown of Oprm1 in the MPOA reduced the number of total play bouts and the frequency of pouncing. Last, RNA sequencing differential gene expression analysis identified genes involved in neuronal signaling with altered expression after Oprm1 knockdown, and identified Egr1 as potentially a key modulator for Oprm1 in the regulation of social play behavior. Altogether, these results show that the MPOA is involved in social play behavior in adolescent male rats and support the hypothesis that the MPOA is part of a conserved neural circuit across vertebrates in which opioids act to govern affiliative, intrinsically rewarded social behaviors.


Assuntos
Proteínas de Transporte/genética , Proteínas de Membrana/genética , Área Pré-Óptica/metabolismo , Receptores Opioides mu/genética , Comportamento Social , Animais , Proteínas de Transporte/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Área Pré-Óptica/crescimento & desenvolvimento , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/metabolismo
19.
Brain Res ; 1748: 147123, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32931818

RESUMO

Long-interspersing element 1 (Line1)-a retrotransposon that comprises ~17% of the human genome and ~24% of the rat genome -is aberrantly expressed in psychiatric disorders such as schizophrenia, bipolar disorder, and Rett syndrome, suggesting it may play an important role in neurodevelopment. Retrotransposons such as Line1 have the ability to self-replicate via reverse transcription and can subsequently be reinserted throughout the genome, potentially increasing genetic diversity. We sought to understand whether early life stress (ELS), a known risk factor for the development of later psychiatric disorders in humans, would affect Line1 expression and DNA copy number. Our study uses a neonatal predator odor exposure (POE) paradigm to model ELS in rats. We found sex- and region-specific increases in both Line1 Open Reading Frame 1 (ORF1) and ORF2 mRNA following POE-induced stress. Interestingly, ELS increased Line1 DNA copy number within the male hippocampus. These data suggest the possibility that early life stress can mobilize Line1 in a sex- and region-specific manner, resulting in genomic heterogeneity between cells in the brain suggesting that some cells may have a different genetic makeup than others resulting in genomic heterogeneity.


Assuntos
Encéfalo/metabolismo , Elementos Nucleotídeos Longos e Dispersos/genética , Estresse Psicológico/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Feminino , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Masculino , Fases de Leitura Aberta/genética , Ratos , Ratos Sprague-Dawley , Fatores de Risco , Fatores Sexuais , Estresse Psicológico/genética
20.
J Neurosci ; 28(28): 7137-42, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18614683

RESUMO

Methyl-CpG-binding protein 2 (MeCP2) binds methylated DNA and recruits corepressor proteins to modify chromatin and alter gene transcription. Mutations of the MECP2 gene can cause Rett syndrome, whereas subtle reductions of MeCP2 expression may be associated with male-dominated social and neurodevelopmental disorders. We report that transiently decreased amygdala Mecp2 expression during a sensitive period of brain sexual differentiation disrupts the organization of sex differences in juvenile social play behavior. Interestingly, neonatal treatment with Mecp2 small interfering RNA within the developing amygdala reduced juvenile social play behavior in males but not females. Reduced Mecp2 expression did not change juvenile sociability or anxiety-like behavior, suggesting that this disruption is associated with subtle behavioral modification. This suggests that Mecp2 may have an overlooked role in the organization of sexually dimorphic behaviors and that male juvenile behavior is particularly sensitive to Mecp2 disruption during this period of development.


Assuntos
Proteína 2 de Ligação a Metil-CpG/metabolismo , Caracteres Sexuais , Comportamento Sexual Animal/fisiologia , Comportamento Social , Adaptação Ocular/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/crescimento & desenvolvimento , Tonsila do Cerebelo/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Ansiedade/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Proteína 2 de Ligação a Metil-CpG/genética , Gravidez , RNA Interferente Pequeno/farmacologia , Ratos , Comportamento Sexual Animal/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA