Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 618(7967): 1006-1016, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37286598

RESUMO

In many species, including mice, female animals show markedly different pup-directed behaviours based on their reproductive state1,2. Naive wild female mice often kill pups, while lactating female mice are dedicated to pup caring3,4. The neural mechanisms that mediate infanticide and its switch to maternal behaviours during motherhood remain unclear. Here, on the basis of the hypothesis that maternal and infanticidal behaviours are supported by distinct and competing neural circuits5,6, we use the medial preoptic area (MPOA), a key site for maternal behaviours7-11, as a starting point and identify three MPOA-connected brain regions that drive differential negative pup-directed behaviours. Functional manipulation and in vivo recording reveal that oestrogen receptor α (ESR1)-expressing cells in the principal nucleus of the bed nucleus of stria terminalis (BNSTprESR1) are necessary, sufficient and naturally activated during infanticide in female mice. MPOAESR1 and BNSTprESR1 neurons form reciprocal inhibition to control the balance between positive and negative infant-directed behaviours. During motherhood, MPOAESR1 and BNSTprESR1 cells change their excitability in opposite directions, supporting a marked switch of female behaviours towards the young.


Assuntos
Infanticídio , Comportamento Materno , Área Pré-Óptica , Animais , Feminino , Camundongos , Lactação , Comportamento Materno/fisiologia , Vias Neurais/fisiologia , Área Pré-Óptica/citologia , Área Pré-Óptica/fisiologia , Tálamo/citologia , Tálamo/fisiologia
2.
Nature ; 596(7873): 553-557, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34381215

RESUMO

Maternal care, including by non-biological parents, is important for offspring survival1-8. Oxytocin1,2,9-15, which is released by the hypothalamic paraventricular nucleus (PVN), is a critical maternal hormone. In mice, oxytocin enables neuroplasticity in the auditory cortex for maternal recognition of pup distress15. However, it is unclear how initial parental experience promotes hypothalamic signalling and cortical plasticity for reliable maternal care. Here we continuously monitored the behaviour of female virgin mice co-housed with an experienced mother and litter. This documentary approach was synchronized with neural recordings from the virgin PVN, including oxytocin neurons. These cells were activated as virgins were enlisted in maternal care by experienced mothers, who shepherded virgins into the nest and demonstrated pup retrieval. Virgins visually observed maternal retrieval, which activated PVN oxytocin neurons and promoted alloparenting. Thus rodents can acquire maternal behaviour by social transmission, providing a mechanism for adapting the brains of adult caregivers to infant needs via endogenous oxytocin.


Assuntos
Aprendizagem , Comportamento Materno/psicologia , Mães/psicologia , Neurônios/metabolismo , Ocitocina/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Abstinência Sexual/psicologia , Ensino , Animais , Feminino , Abrigo para Animais , Tamanho da Ninhada de Vivíparos , Camundongos , Comportamento de Nidação , Plasticidade Neuronal
3.
Proc Natl Acad Sci U S A ; 116(45): 22821-22832, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31636210

RESUMO

Infant maltreatment increases vulnerability to physical and mental disorders, yet specific mechanisms embedded within this complex infant experience that induce this vulnerability remain elusive. To define critical features of maltreatment-induced vulnerability, rat pups were reared from postnatal day 8 (PN8) with a maltreating mother, which produced amygdala and hippocampal deficits and decreased social behavior at PN13. Next, we deconstructed the maltreatment experience to reveal sufficient and necessary conditions to induce this phenotype. Social behavior and amygdala deficits (volume, neurogenesis, c-Fos, local field potential) required combined chronic high corticosterone and maternal presence (not maternal behavior). Hippocampal deficits were induced by chronic high corticosterone regardless of social context. Causation was shown by blocking corticosterone during maltreatment and suppressing amygdala activity during social behavior testing. These results highlight (1) that early life maltreatment initiates multiple pathways to pathology, each with distinct causal mechanisms and outcomes, and (2) the importance of social presence on brain development.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Hipocampo/fisiopatologia , Mães/psicologia , Comportamento Social , Estresse Fisiológico , Animais , Corticosterona/administração & dosagem , Corticosterona/sangue , Feminino , Ratos
4.
Annu Rev Clin Psychol ; 15: 345-369, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30786246

RESUMO

Children's development is largely dependent on caregiving; when caregiving is disrupted, children are at increased risk for numerous poor outcomes, in particular psychopathology. Therefore, determining how caregivers regulate children's affective neurobiology is essential for understanding psychopathology etiology and prevention. Much of the research on affective functioning uses fear learning to map maturation trajectories, with both rodent and human studies contributing knowledge. Nonetheless, as no standard framework exists through which to interpret developmental effects across species, research often remains siloed, thus contributing to the current therapeutic impasse. Here, we propose a developmental ecology framework that attempts to understand fear in the ecological context of the child: their relationship with their parent. By referring to developmental goals that are shared across species (to attach to, then, ultimately, separate from the parent), this framework provides a common grounding from which fear systems and their dysfunction can be understood, thus advancing research on psychopathologies and their treatment.


Assuntos
Comportamento Animal/fisiologia , Desenvolvimento Infantil/fisiologia , Medo/fisiologia , Aprendizagem/fisiologia , Apego ao Objeto , Relações Pais-Filho , Animais , Criança , Humanos
5.
Dev Psychopathol ; 31(2): 399-418, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29606185

RESUMO

Children reared in impoverished environments are at risk for enduring psychological and physical health problems. Mechanisms by which poverty affects development, however, remain unclear. To explore one potential mechanism of poverty's impact on social-emotional and cognitive development, an experimental examination of a rodent model of scarcity-adversity was conducted and compared to results from a longitudinal study of human infants and families followed from birth (N = 1,292) who faced high levels of poverty-related scarcity-adversity. Cross-species results supported the hypothesis that altered caregiving is one pathway by which poverty adversely impacts development. Rodent mothers assigned to the scarcity-adversity condition exhibited decreased sensitive parenting and increased negative parenting relative to mothers assigned to the control condition. Furthermore, scarcity-adversity reared pups exhibited decreased developmental competence as indicated by disrupted nipple attachment, distress vocalization when in physical contact with an anesthetized mother, and reduced preference for maternal odor with corresponding changes in brain activation. Human results indicated that scarcity-adversity was inversely correlated with sensitive parenting and positively correlated with negative parenting, and that parenting fully mediated the association of poverty-related risk with infant indicators of developmental competence. Findings are discussed from the perspective of the usefulness of bidirectional-translational research to inform interventions for at-risk families.


Assuntos
Modelos Animais , Poder Familiar/psicologia , Pobreza/psicologia , Animais , Feminino , Estudos Longitudinais , Masculino , Ratos , Meio Social
6.
Proc Natl Acad Sci U S A ; 112(3): 881-6, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25561533

RESUMO

Children form a strong attachment to their caregiver--even when that caretaker is abusive. Paradoxically, despite the trauma experienced within this relationship, the child develops a preference for trauma-linked cues--a phenomenon known as trauma bonding. Although infant trauma compromises neurobehavioral development, the mechanisms underlying the interaction between infant trauma bonding (i.e., learned preference for trauma cues) and the long-term effects of trauma (i.e., depressive-like behavior, amygdala dysfunction) are unknown. We modeled infant trauma bonding by using odor-shock conditioning in rat pups, which engages the attachment system and produces a life-long preference for the odor that was paired with shock. In adulthood, this trauma-linked odor rescues depressive-like behavior and amygdala dysfunction, reduces corticosterone (CORT) levels, and exerts repair-related changes at the molecular level. Amygdala microarray after rescue implicates serotonin (5-HT) and glucocorticoids (GCs), and a causal role was verified through microinfusions. Blocking amygdala 5-HT eliminates the rescue effect; increasing amygdala 5-HT and blocking systemic CORT mimics it. Our findings suggest that infant trauma cues share properties with antidepressants and safety signals and provide insight into mechanisms by which infant trauma memories remain powerful throughout life.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Comportamento , Corticosterona/metabolismo , Memória , Serotonina/metabolismo , Ferimentos e Lesões/psicologia , Adulto , Tonsila do Cerebelo/metabolismo , Humanos , Lactente
7.
Learn Mem ; 24(3): 115-122, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28202715

RESUMO

The updating of a memory is triggered whenever it is reactivated and a mismatch from what is expected (i.e., prediction error) is detected, a process that can be unraveled through the memory's sensitivity to protein synthesis inhibitors (i.e., reconsolidation). As noted in previous studies, in Pavlovian threat/aversive conditioning in adult rats, prediction error detection and its associated protein synthesis-dependent reconsolidation can be triggered by reactivating the memory with the conditioned stimulus (CS), but without the unconditioned stimulus (US), or by presenting a CS-US pairing with a different CS-US interval than during the initial learning. Whether similar mechanisms underlie memory updating in the young is not known. Using similar paradigms with rapamycin (an mTORC1 inhibitor), we show that preweaning rats (PN18-20) do form a long-term memory of the CS-US interval, and detect a 10-sec versus 30-sec temporal prediction error. However, the resulting updating/reconsolidation processes become adult-like after adolescence (PN30-40). Our results thus show that while temporal prediction error detection exists in preweaning rats, specific infant-type mechanisms are at play for associative learning and memory.


Assuntos
Envelhecimento/fisiologia , Aprendizagem da Esquiva/fisiologia , Condicionamento Clássico/fisiologia , Memória/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Sinais (Psicologia) , Relação Dose-Resposta a Droga , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Medo/efeitos dos fármacos , Feminino , Reação de Congelamento Cataléptica/efeitos dos fármacos , Reação de Congelamento Cataléptica/fisiologia , Imunossupressores/farmacologia , Masculino , Memória/efeitos dos fármacos , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Sirolimo/farmacologia , Paladar/efeitos dos fármacos , Paladar/fisiologia
8.
J Neurosci ; 36(25): 6634-50, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27335397

RESUMO

UNLABELLED: A major component of perception is hedonic valence: perceiving stimuli as pleasant or unpleasant. Here, we used early olfactory experiences that shape odor preferences and aversions to explore developmental plasticity in circuits mediating odor hedonics. We used 2-deoxyglucose autoradiographic mapping of neural activity to identify circuits differentially activated by biologically relevant preferred and avoided odors across rat development. We then further probed this system by increasing or decreasing hedonic value. Using both region of interest and functional connectivity analyses, we identified regions within primary olfactory, amygdala/hippocampal, and prefrontal cortical networks that were activated differentially by maternal and male odors. Although some activated regions remained stable across development (postnatal days 7-23), there was a developmental emergence of others that resulted in an age-dependent elaboration of hedonic-response-specific circuitry despite stable behavioral responses (approach/avoidance) to the odors across age. Hedonic responses to these biologically important odors were modified through diet suppression of the maternal odor and co-rearing with a male. This allowed assessment of hedonic circuits in isolation of the specific odor quality and/or intensity. Early experience significantly modified odor-evoked circuitry in an age-dependent manner. For example, co-rearing with a male, which induced pup attraction to male odor, reduced activity in amygdala regions normally activated by the unfamiliar avoided male odor, making this region more consistent with maternal odor. Understanding the development of odor hedonics, particularly within the context of altered early life experience, provides insight into the development of sensory processes, food preferences, and the formation of social affiliations, among other behaviors. SIGNIFICANCE STATEMENT: Odor hedonic valence controls approach-avoidance behaviors, but also modulates ongoing behaviors ranging from food preferences and social affiliation with the caregiver to avoidance of predator odors. Experiences can shape hedonic valence. This study explored brain circuitry involved in odor hedonic encoding throughout development using maternal and predator odors and assessed the effects of early life experience on odor hedonic encoding by increasing/decreasing the hedonic value of these odors. Understanding the role of changing brain circuitry during development and its impact on behavioral function is critical for understanding sensory processing across development. These data converge with exciting literature on the brain's hedonic network and highlight the significant role of early life experience in shaping the neural networks of highly biologically relevant stimuli.


Assuntos
Anedonia/fisiologia , Encéfalo/fisiologia , Odorantes , Condutos Olfatórios/fisiologia , Percepção Olfatória/fisiologia , Olfato , Animais , Animais Recém-Nascidos , Autorradiografia , Aprendizagem da Esquiva/fisiologia , Encéfalo/diagnóstico por imagem , Desoxiglucose/metabolismo , Emoções/fisiologia , Feminino , Masculino , Condutos Olfatórios/diagnóstico por imagem , Ratos , Ratos Long-Evans
9.
Neurobiol Learn Mem ; 143: 49-58, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27826033

RESUMO

What an animal needs to learn to survive is altered dramatically as they change from dependence on the parent for protection to independence and reliance on self-defense. This transition occurs in most altricial animals, but our understanding of the behavioral neurobiology has mostly relied on the infant rat. The transformation from dependence to independence occurs over three weeks in pups and is accompanied by complex changes in responses to both natural and learned threats and the supporting neural circuitry. Overall, in early life, the threat system is quiescent and learning is biased towards acquiring attachment related behaviors to support attachment to the caregiver and proximity seeking. Caregiver-associated cues learned in infancy have the ability to provide a sense of safety throughout lifetime. This attachment/safety system is activated by learning involving presumably pleasurable stimuli (food, warmth) but also painful stimuli (tailpinch, moderate shock). At about the midway point to independence, pups begin to have access to the adult-like amygdala-dependent threat system and amygdala-dependent responses to natural dangers such as predator odors. However, pups have the ability to switch between the infant and adult-like system, which is controlled by maternal presence and modification of stress hormones. Specifically, if the pup is alone, it will learn fear but if with the mother it will learn attachment (10-15days of age). As pups begin to approach weaning, pups lose access to the attachment system and rely only on the amygdala-dependent threat system. However, pups learning system is complex and exhibits flexibility that enables the mother to override the control of the attachment circuit, since newborn pups may acquire threat responses from the mother expressing fear in their presence. Together, these data suggest that the development of pups' threat learning system is not only dependent upon maturation of the amygdala, but it is also exquisitely controlled by the environment. Most notably the mother can switch pup learning between attachment to threat learning in a moment's notice. This enables the mother to navigate pup's learning about the world and what is threatening and what is safe.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Aprendizagem/fisiologia , Apego ao Objeto , Tonsila do Cerebelo/crescimento & desenvolvimento , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Humanos , Odorantes , Estresse Psicológico
10.
Stress ; 20(5): 421-448, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28617197

RESUMO

The immediate and long-term effects of exposure to early life stress (ELS) have been documented in humans and animal models. Even relatively brief periods of stress during the first 10 days of life in rodents can impact later behavioral regulation and the vulnerability to develop adult pathologies, in particular an impairment of cognitive functions and neurogenesis, but also modified social, emotional, and conditioned fear responses. The development of preclinical models of ELS exposure allows the examination of mechanisms and testing of therapeutic approaches that are not possible in humans. Here, we describe limited bedding and nesting (LBN) procedures, with models that produce altered maternal behavior ranging from fragmentation of care to maltreatment of infants. The purpose of this paper is to discuss important issues related to the implementation of this chronic ELS procedure and to describe some of the most prominent endpoints and consequences, focusing on areas of convergence between laboratories. Effects on the hypothalamic-pituitary adrenal (HPA) axis, gut axis and metabolism are presented in addition to changes in cognitive and emotional functions. Interestingly, recent data have suggested a strong sex difference in some of the reported consequences of the LBN paradigm, with females being more resilient in general than males. As both the chronic and intermittent variants of the LBN procedure have profound consequences on the offspring with minimal external intervention from the investigator, this model is advantageous ecologically and has a large translational potential. In addition to the direct effect of ELS on neurodevelopmental outcomes, exposure to adverse early environments can also have intergenerational impacts on mental health and function in subsequent generation offspring. Thus, advancing our understanding of the effect of ELS on brain and behavioral development is of critical concern for the health and wellbeing of both the current population, and for generations to come.


Assuntos
Maus-Tratos Infantis , Cognição , Emoções , Comportamento Materno , Comportamento de Nidação , Estresse Psicológico/psicologia , Tecido Adiposo Branco/metabolismo , Animais , Animais Recém-Nascidos , Roupas de Cama, Mesa e Banho , Comportamento Animal , Epigênese Genética , Feminino , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Recém-Nascido , Masculino , Modelos Animais , Neurogênese , Sistema Hipófise-Suprarrenal/metabolismo , Reprodutibilidade dos Testes , Resiliência Psicológica , Roedores , Fatores Sexuais , Estresse Psicológico/metabolismo
11.
Learn Mem ; 23(10): 556-66, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27634146

RESUMO

As altricial infants gradually transition to adults, their proximate environment changes. In three short weeks, pups transition from a small world with the caregiver and siblings to a complex milieu rich in dangers as their environment expands. Such contrasting environments require different learning abilities and lead to distinct responses throughout development. Here, we will review some of the learned fear conditioned responses to threats in rats during their ontogeny, including behavioral and physiological measures that permit the assessment of learning and its supporting neurobiology from infancy through adulthood. In adulthood, odor-shock conditioning produces robust fear learning to the odor that depends upon the amygdala and related circuitry. Paradoxically, this conditioning in young pups fails to support fear learning and supports approach learning to the odor previously paired with shock. This approach learning is mediated by the infant attachment network that does not include the amygdala. During the age range when pups transition from the infant to the adult circuit (10-15 d old), pups have access to both networks: odor-shock conditioning in maternal presence uses the attachment circuit but the adult amygdala-dependent circuit when alone. However, throughout development (as young as 5 d old) the attachment associated learning can be overridden and amygdala-dependent fear learning supported, if the mother expresses fear in the presence of the pup. This social modulation of the fear permits the expression of defense reactions in life threatening situations informed by the caregiver but prevents the learning of the caregiver itself as a threat.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Medo/fisiologia , Aprendizagem/fisiologia , Animais , Ratos
12.
Dev Psychobiol ; 56(8): 1626-34, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24771610

RESUMO

Childhood maltreatment is associated with adverse brain development and later life psychiatric disorders, with maltreatment from the caregiver inducing a particular vulnerability to later life psychopathologies. Here we review two complementary rodent models of early life abuse, which are used to examine the infant response to trauma within attachment and the developmental trajectories that lead to later life neurobehavioral deficits. These rodent models include being reared with an abusive mother, and a more controlled attachment-learning paradigm using odor-shock conditioning to produce a new maternal odor. In both of these rodent models, pups learn a strong attachment and preference to the maternal odor. However, both models produce similar enduring neurobehavioral deficits, which emerge with maturation. Importantly, cues associated with our models of abuse serve as paradoxical safety signals, by normalizing enduring neurobehavioral deficits following abuse. Here we review these models and explore implications for human interventions for early life maltreatment.


Assuntos
Animais Recém-Nascidos , Maus-Tratos Infantis/psicologia , Modelos Animais de Doenças , Comportamento Materno/fisiologia , Apego ao Objeto , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/fisiologia , Animais Recém-Nascidos/psicologia , Humanos , Recém-Nascido , Comportamento Materno/psicologia , Ratos
13.
Dev Psychobiol ; 56(8): 1635-50, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25290865

RESUMO

Early-life caregiving shapes the architecture and function of the developing brain. The fact that the infant-caregiver relationship is critically important for infant functioning across all altricial species, and that the anatomical circuits supporting emotional functioning are highly preserved across different species, suggests that the results of studies examining the role of early adversity and emotional functioning should be translatable across species. Here we present findings from four different research laboratories, using three different species, which have converged on a similar finding: adversity accelerates the developmental trajectory of amygdala-prefrontal cortex (PFC) development and modifies emotional behaviors. First, a rodent model of attachment learning associated with adversity is presented showing precocial disruption of attachment learning and emergence of heightened fear learning and emotionality. Second, a model of infant-mother separation is presented in which early adversity is shown to accelerate the developmental emergence of adult-like fear retention and extinction. Third, a model of early life adversity in Rhesus monkeys is presented in which a naturally occurring variation in maternal-care (abuse) is shown to alter the functioning of emotion circuits. Finally, a human model of maternal deprivation is presented in which children born into orphanages and then adopted abroad exhibit aberrant development of emotion circuits. The convergence of these cross-species studies on early life adversity suggests that adversity targets the amygdala and PFC and has immediate impact on infant behavior with the caregiver, and emotional reactions to the world. These results provide insight into mechanisms responsible for caregiver induced mental health trajectory alterations.


Assuntos
Tonsila do Cerebelo/crescimento & desenvolvimento , Animais Recém-Nascidos , Maus-Tratos Infantis , Modelos Animais de Doenças , Medo/fisiologia , Privação Materna , Apego ao Objeto , Córtex Pré-Frontal/crescimento & desenvolvimento , Tonsila do Cerebelo/fisiopatologia , Animais , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/metabolismo , Animais Recém-Nascidos/fisiologia , Humanos , Lactente , Macaca mulatta , Córtex Pré-Frontal/fisiopatologia , Ratos
14.
Neurosci Biobehav Rev ; 158: 105567, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38309498

RESUMO

Prenatal alcohol exposure is the leading nongenetic cause of human intellectual impairment. The long-term impacts of prenatal alcohol exposure on health and well-being are diverse, including neuropathology leading to behavioral, cognitive, and emotional impairments. Additionally negative effects also occur on the physiological level, such as the endocrine, cardiovascular, and immune systems. Among these diverse impacts is sleep disruption. In this review, we describe how prenatal alcohol exposure affects sleep, and potential mechanisms of those effects. Furthermore, we outline the evidence that sleep disruption across the lifespan may be a mediator of some cognitive and behavioral impacts of developmental alcohol exposure, and thus may represent a promising target for treatment.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Efeitos Tardios da Exposição Pré-Natal , Feminino , Humanos , Gravidez , Transtornos do Espectro Alcoólico Fetal/etiologia , Etanol/efeitos adversos , Sono
15.
J Neurosci ; 32(22): 7758-65, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649253

RESUMO

Abuse during early life, especially from the caregiver, increases vulnerability to develop later-life psychopathologies such as depression. Although signs of depression are typically not expressed until later life, signs of dysfunctional social behavior have been found earlier. How infant abuse alters the trajectory of brain development to produce pathways to pathology is not completely understood. Here we address this question using two different but complementary rat models of early-life abuse from postnatal day 8 (P8) to P12: a naturalistic paradigm, where the mother is provided with insufficient bedding for nest building; and a more controlled paradigm, where infants undergo olfactory classical conditioning. Amygdala neural assessment (c-Fos), as well as social behavior and forced swim tests were performed at preweaning (P20) and adolescence (P45). Our results show that both models of early-life abuse induce deficits in social behavior, even during the preweaning period; however, depressive-like behaviors were observed only during adolescence. Adolescent depressive-like behavior corresponds with an increase in amygdala neural activity in response to forced swim test. A causal relationship between the amygdala and depressive-like behavior was suggested through amygdala temporary deactivation (muscimol infusions), which rescued the depressive-like behavior in the forced swim test. Our results indicate that social behavior deficits in infancy could serve as an early marker for later psychopathology. Moreover, the implication of the amygdala in the ontogeny of depressive-like behaviors in infant abused animals is an important step toward understanding the underlying mechanisms of later-life mental disease associated with early-life abuse.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Transtorno Depressivo/etiologia , Deficiências do Desenvolvimento/etiologia , Transtornos do Comportamento Social/complicações , Transtornos do Comportamento Social/patologia , Adolescente , Fatores Etários , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Modelos Animais de Doenças , Feminino , Agonistas de Receptores de GABA-A/farmacologia , Humanos , Masculino , Comportamento Materno/psicologia , Privação Materna , Muscimol/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Condutos Olfatórios/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Long-Evans , Isolamento Social/psicologia , Natação/psicologia , Vocalização Animal/fisiologia
16.
Curr Biol ; 33(20): R1049-R1051, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37875078

RESUMO

Developmental neural activity organizes sensory system development. New evidence in mice suggests postnatal olfactory bulb activity also modulates development of the structure and function of hippocampal-cortical circuits. Reducing cell-specific olfactory bulb output during an infant sensitive period impairs later-life cognition.


Assuntos
Neurociências , Olfato , Humanos , Camundongos , Animais , Bulbo Olfatório , Cognição , Hipocampo , Odorantes
17.
Neurosci Biobehav Rev ; 151: 105249, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37257712

RESUMO

Infant survival relies on rapid identification, remembering and behavioral responsiveness to caregivers' sensory cues. While neural circuits supporting infant attachment learning have largely remained elusive in children, use of invasive techniques has uncovered some of its features in rodents. During a 10-day sensitive period from birth, newborn rodents associate maternal odors with maternal pleasant or noxious thermo-tactile stimulation, which gives rise to a preference and approach behavior towards these odors, and blockade of avoidance learning. Here we review the neural circuitry supporting this neonatal odor learning, unique compared to adults, focusing specifically on the early roles of neurotransmitters such as glutamate, GABA (Gamma-AminoButyric Acid), serotonin, dopamine and norepinephrine, in the olfactory bulb, the anterior piriform cortex and amygdala. The review highlights the importance of deepening our knowledge of age-specific infant brain neurotransmitters and behavioral functioning that can be translated to improve the well-being of children during typical development and aid in treatment during atypical development in childhood clinical practice, and the care during rearing of domestic animals.


Assuntos
Odorantes , Bulbo Olfatório , Ratos , Animais , Animais Recém-Nascidos , Ratos Long-Evans , Bulbo Olfatório/fisiologia , Aprendizagem da Esquiva , Neurotransmissores , Olfato/fisiologia
18.
PLoS One ; 18(11): e0290871, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37972112

RESUMO

BACKGROUND: In the short term, parental presence while a human infant is in pain buffers the immediate pain responses, although emerging evidence suggests repeated social buffering of pain may have untoward long-term effects. METHODS/FINDING: To explore the short- and long-term impacts of social buffering of pain, we first measured the infant rat pup's [postnatal day (PN) 8, or 12] response to mild tail shock with the mother present compared to shock alone or no shock. Shock with the mother reduced pain-related behavioral activation and USVs of pups at both ages and reduced Fos expression in the periaqueductal gray, hypothalamic paraventricular nucleus, and the amygdala at PN12 only. At PN12, shock with the mother compared to shock alone differentially regulated expression of several hundred genes related to G-protein-coupled receptors (GPCRs) and neural development, whereas PN8 pups showed a less robust and less coherent expression pattern. In a second set of experiments, pups were exposed to daily repeated Shock-mother pairings (or controls) at PN5-9 or PN10-14 (during and after pain sensitive period, respectively) and long-term outcome assessed in adults. Shock+mother pairing at PN5-9 reduced adult carrageenan-induced thermal hyperalgesia and reduced Fos expression, but PN10-14 pairings had minimal impact. The effect of infant treatment on adult affective behavior showed a complex treatment by age dependent effect. Adult social behavior was decreased following Shock+mother pairings at both PN5-9 and PN10-14, whereas shock alone had no effect. Adult fear responses to a predator odor were decreased only by PN10-14 treatment and the infant Shock alone and Shock+mother did not differ. CONCLUSIONS/SIGNIFICANCE: Overall, integrating these results into our understanding of long-term programming by repeated infant pain experiences, the data suggest that pain experienced within a social context impacts infant neurobehavioral responses and initiates an altered developmental trajectory of pain and affect processing that diverges from experiencing pain alone.


Assuntos
Encéfalo , Mães , Feminino , Humanos , Animais , Ratos , Lactente , Encéfalo/fisiologia , Odorantes , Comportamento Social , Dor/metabolismo , Animais Recém-Nascidos
19.
Front Neurosci ; 17: 1267542, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38033546

RESUMO

Developmental exposure to ethanol is a leading cause of cognitive, emotional and behavioral problems, with fetal alcohol spectrum disorder (FASD) affecting more than 1:100 children. Recently, comorbid sleep deficits have been highlighted in these disorders, with sleep repair a potential therapeutic target. Animal models of FASD have shown non-REM (NREM) sleep fragmentation and slow-wave oscillation impairments that predict cognitive performance. Here we use a mouse model of perinatal ethanol exposure to explore whether reduced sleep pressure may contribute to impaired NREM sleep, and compare the function of a brain network reported to be impacted by insomnia-the Salience network-in developmental ethanol-exposed mice with sleep-deprived, saline controls. Mice were exposed to ethanol or saline on postnatal day 7 (P7) and allowed to mature to adulthood for testing. At P90, telemetered cortical recordings were made for assessment of NREM sleep in home cage before and after 4 h of sleep deprivation to assess basal NREM sleep and homeostatic NREM sleep response. To assess Salience network functional connectivity, mice were exposed to the 4 h sleep deprivation period or left alone, then immediately sacrificed for immunohistochemical analysis of c-Fos expression. The results show that developmental ethanol severely impairs both normal rebound NREM sleep and sleep deprivation induced increases in slow-wave activity, consistent with reduced sleep pressure. Furthermore, the Salience network connectome in rested, ethanol-exposed mice was most similar to that of sleep-deprived, saline control mice, suggesting a sleep deprivation-like state of Salience network function after developmental ethanol even without sleep deprivation.

20.
Dev Neurosci ; 34(2-3): 101-14, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22571921

RESUMO

Survival of altricial infants depends on attachment to the caregiver - a process that requires infants to identify, learn, remember, and approach their attachment figure. Here we review the neurobiology of attachment in infant rats where learning about the caregiver is supported by a specialized attachment neural circuitry to promote the infant-caregiver relationship. Specifically, the attachment circuit relies on infants acquiring learned preferences to the maternal odor, and this behavior is supported by the hyperfunctioning locus coeruleus and generous amounts of norepinephrine to produce experience-induced changes in the olfactory bulb and anterior piriform cortex. Infants also possess a reduced ability to acquire learned aversions or fear, and this behavior is facilitated through attenuated amygdala plasticity to block fear learning. Presumably, this attachment circuitry constrains the infant animal to express only learned preferences regardless of the quality of care received. As pups mature, and begin to travel in and out of the nest, the specialized attachment learning becomes contextually confined to when pups are with the mother. Thus, when outside the nest, these older pups show learning more typical of adult learning, presumably to prepare for independent life outside the nest. The quality of attachment can alter this circuitry, with early life stress prematurely terminating the pups' access to the attachment system through premature functional activation of the amygdala. Overall, the attachment circuit appears to have a dual function: to keep pups close to the caregiver but also to shape pups' behavior to match the environment and define long-term emotion and cognition.


Assuntos
Medo/fisiologia , Aprendizagem/fisiologia , Comportamento Materno/fisiologia , Apego ao Objeto , Bulbo Olfatório/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Odorantes , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA