Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Neurotoxicol Teratol ; 103: 107353, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38648864

RESUMEN

Aversive caregiving in early life is a risk factor for aberrant brain and behavioral development. This outcome is related to epigenetic dysregulation of the brain-derived neurotrophic factor (Bdnf) gene. The Bdnf gene encodes for BDNF, a neurotrophin involved in early brain development, neural plasticity, learning, and memory. Recent work suggests that exercise may be neuroprotective in part by supporting BDNF protein and gene expression, making it an exciting target for therapeutic interventions. To our knowledge, exercise has never been studied as a therapeutic intervention in preclinical rodent models of caregiver maltreatment. To that end, the current study investigated the effect of an adult voluntary wheel running intervention on Bdnf methylation and expression in the prefrontal cortex of rats who experienced aversive caregiving in infancy. We employed a rodent model (Long Evans rats) wherein rat pups experienced intermittent caregiver-induced stress from postnatal days 1-7 and were given voluntary access to a running wheel (except in the control condition) from postnatal days 70-90 as a young adulthood treatment intervention. Our results indicate that maltreatment and exercise affect Bdnf gene methylation in an exon, CG site, and sex-specific manner. Here we add to a growing body of evidence of the ability for our experiences, including exercise, to permeate the brain. Keywords: Early life stress, Bdnf, exercise, prefrontal cortex.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Metilación de ADN , Condicionamiento Físico Animal , Corteza Prefrontal , Ratas Long-Evans , Estrés Psicológico , Animales , Corteza Prefrontal/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Masculino , Condicionamiento Físico Animal/fisiología , Femenino , Estrés Psicológico/metabolismo , Estrés Psicológico/genética , Ratas , Epigenoma , Epigénesis Genética
2.
Dev Psychobiol ; 66(2): e22466, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38388192

RESUMEN

In early development, the spinal cord in healthy or disease states displays remarkable activity-dependent changes in plasticity, which may be in part due to the increased activity of brain derived neurotrophic factor (BDNF). Indeed, BDNF delivery has been efficacious in partially ameliorating many of the neurobiological and behavioral consequences of spinal cord injury (SCI), making elucidating the role of BDNF in the normative developing and injured spinal cord a critical research focus. Recent work in our laboratory provided evidence for aberrant global and locus-specific epigenetic changes in methylation of the Bdnf gene as a consequence of SCI. In the present study, animals underwent thoracic lesions on P1, with cervical and lumbar tissue being later collected on P7, P14, and P21. Levels of Bdnf expression and methylation (exon IX and exon IV), in addition to global methylation levels were quantified at each timepoint. Results indicated locus-specific reductions of Bdnf expression that was accompanied by a parallel increase in methylation caudal to the injury site, with animals displaying increased Bdnf expression at the P14 timepoint. Together, these findings suggest that epigenetic activity of the Bdnf gene may act as biomarker in the etiology and intervention effort efficacy following SCI.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Traumatismos de la Médula Espinal , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/metabolismo , Epigénesis Genética
3.
Cells ; 12(7)2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-37048047

RESUMEN

A total of 1 in 20 infants born annually are exposed to alcohol prenatally, which disrupts neurodevelopment and results in several disorders categorized under the umbrella term Fetal Alcohol Spectrum Disorders (FASD). Children and adolescents affected by FASD exhibit delayed maturation of cerebral white matter, which contributes to deficits in executive function, visuospatial processing, sensory integration, and interhemispheric communication. Research using animal models of FASD have uncovered that oligoglia proliferation, differentiation, and survival are vulnerable to alcohol teratogenesis in the male brain due in part to the activation of the neuroimmune system during gestation and infancy. A comprehensive investigation of prenatal alcohol exposure on white matter development in the female brain is limited. This study demonstrated that the number of mature oligodendrocytes and the production of myelin basic protein were reduced first in the female corpus callosum following alcohol exposure in a rat model of FASD. Analysis of myelin-related genes confirmed that myelination occurs earlier in the female corpus callosum compared to their counterparts, irrespective of postnatal treatment. Moreover, dysregulated oligodendrocyte number and myelin basic protein production was observed in the male and female FASD brain in adolescence. Targeted interventions that support white matter development in FASD-affected youth are nonexistent. The capacity for an adolescent exercise intervention to upregulate corpus callosum myelination was evaluated: we discovered that volunteer exercise increases the number of mature oligodendrocytes in alcohol-exposed female rats. This study provides critical evidence that oligoglia differentiation is difficult but not impossible to induce in the female FASD brain in adolescence following a behavioral intervention.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Efectos Tardíos de la Exposición Prenatal , Sustancia Blanca , Humanos , Femenino , Masculino , Ratas , Embarazo , Animales , Cuerpo Calloso , Proteína Básica de Mielina , Encéfalo , Etanol/toxicidad
4.
Int J Mol Sci ; 23(19)2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-36233029

RESUMEN

Early life stress (ELS) encompasses exposure to aversive experiences during early development, such as neglect or maltreatment. Animal and human studies indicate that ELS has maladaptive effects on brain development, leaving individuals more vulnerable to developing behavioral and neuropsychiatric disorders later in life. This result occurs in part to disruptions in Brain derived neurotrophic factor (Bdnf) gene regulation, which plays a vital role in early neural programming and brain health in adulthood. A potential treatment mechanism to reverse the effects of ELS on Bdnf expression is aerobic exercise due to its neuroprotective properties and positive impact on Bdnf expression. Aerobic exercise opens the door to exciting and novel potential treatment strategies because it is a behavioral intervention readily and freely available to the public. In this review, we discuss the current literature investigating the use of exercise interventions in animal models of ELS to reverse or mitigate ELS-induced changes in Bdnf expression. We also encourage future studies to investigate sensitive periods of exercise exposure, as well as sufficient duration of exposure, on epigenetic and behavioral outcomes to help lead to standardized practices in the exercise intervention field.


Asunto(s)
Experiencias Adversas de la Infancia , Factor Neurotrófico Derivado del Encéfalo , Adulto , Animales , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ejercicio Físico/fisiología , Terapia por Ejercicio , Humanos , Estrés Psicológico
5.
Dev Psychobiol ; 64(3): e22231, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35312054

RESUMEN

Exposure to adversity in early development has powerful and potentially lasting consequences on behavior. Previous work in our laboratory using female Long-Evans rats has demonstrated that exposure to early-life maltreatment manifests into alterations in dam behavior, including a perpetuation of the maltreatment phenotype. These observed behavioral changes coincide with changes in epigenetic activity in the prefrontal cortex (PFC). Further, treating dams with a chromatin modifying agent (Zebularine) normalizes methylation and maltreatment phenotypes, suggesting a link between epigenetic programming and phenotypic outcomes. Here, we sought to investigate if administration of a chromatin modifying agent concurrent with the experience of maltreatment normalizes epigenetic activity associated with maltreatment and alters behavioral trajectories. Administration of valproic acid (VPA) transiently lowered levels of global DNA methylation in the PFC, regardless of exposure to nurturing care or maltreatment. When VPA-exposed animals reached adulthood, they engaged in more adverse behaviors toward their offspring. These data provide further evidence linking epigenetic changes in the developing brain with effects on behavior.


Asunto(s)
Metilación de ADN , Ácido Valproico , Adulto , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Humanos , Conducta Materna , Ratas , Ratas Long-Evans , Ácido Valproico/farmacología
6.
Front Behav Neurosci ; 15: 786234, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34924972

RESUMEN

Early-life adversity (ELA), often clinically referred to as "adverse childhood experiences (ACE)," is the exposure to stress-inducing events in childhood that can result in poor health outcomes. ELA negatively affects neurodevelopment in children and adolescents resulting in several behavioral deficits and increasing the risk of developing a myriad of neuropsychiatric disorders later in life. The neurobiological mechanisms by which ELA alters neurodevelopment in childhood have been the focus of numerous reviews. However, a comprehensive review of the mechanisms affecting adolescent neurodevelopment (i.e., synaptic pruning and myelination) is lacking. Synaptic pruning and myelination are glia-driven processes that are imperative for brain circuit refinement during the transition from adolescence to adulthood. Failure to optimize brain circuitry between key brain structures involved in learning and memory, such as the hippocampus and prefrontal cortex, leads to the emergence of maladaptive behaviors including increased anxiety or reduced executive function. As such, we review preclinical and clinical literature to explore the immediate and lasting effects of ELA on brain circuit development and refinement. Finally, we describe a number of therapeutic interventions best-suited to support adolescent neurodevelopment in children with a history of ELA.

7.
Front Hum Neurosci ; 14: 594244, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324186

RESUMEN

Child maltreatment not only leads to epigenetic changes, but also increases the risk of related behavioral deficits and mental disorders. These issues presumably are most closely associated with epigenetic changes in the brain, but epigenetic changes in peripheral tissues like blood are often examined instead, due to their accessibility. As such, the reliability of using the peripheral epigenome as a proxy for that of the brain is imperative. Previously, our lab has found aberrant methylation at the Brain-derived neurotrophic factor (Bdnf) gene in the prefrontal cortex of rats following aversive caregiving. The current study examined whether aversive caregiving alters Bdnf DNA methylation in the blood compared to the prefrontal cortex. It was revealed that DNA methylation associated with adversity increased in both tissues, but this methylation was not correlated between tissues. These findings indicate that group trends in Bdnf methylation between blood and the brain are comparable, but variation exists among individual subjects.

9.
Dev Psychopathol ; 32(4): 1486-1494, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31854285

RESUMEN

Maltreatment during development is associated with epigenetic changes to the genome. Enhancing caregiving may mitigate these effects. Attachment and Biobehavioral Catch-Up (ABC) is an intervention that has been shown to improve parent-child relationships and a variety of biological and behavioral outcomes among children that are involved in Child Protective Services. This preliminary study, using a small sample size, explored whether children who received ABC exhibit different methylation patterns than those who received a control intervention. The participants included 23 children aged 6-21 months who were randomized to receive ABC (n = 12) or a control intervention (n = 11). While the children displayed similar methylation patterns preintervention, DNA methylation varied between the ABC and control groups at 14,828 sites postintervention. Functional pathway analyses indicated that these differences were associated with gene pathways that are involved in cell signaling, metabolism, and neuronal development. This study is one of the first to explore parenting intervention effects on children's DNA methylation at the whole genome level in infancy. These preliminary findings provide a basis for hypothesis generation in further research with larger-scale studies regarding the malleability of epigenetic states that are associated with maltreatment.


Asunto(s)
Metilación de ADN , Apego a Objetos , Relaciones Padres-Hijo , Niño , Maltrato a los Niños , Servicios de Protección Infantil , Humanos , Lactante , Responsabilidad Parental
10.
Infant Behav Dev ; 57: 101381, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31557646

RESUMEN

Although the importance of epigenetic mechanisms in behavioral development has been gaining attention in recent years, research has largely focused on the brain. To our knowledge, no studies to date have investigated epigenetic changes in the developing spinal cord to determine the dynamic manner in which the spinal epigenome may respond to environmental input during behavioral development. Animal studies demonstrate that spinal cord plasticity is heightened during early development, is somewhat preserved following neonatal transection, and that spinal injured animals are responsive to sensory feedback. Because epigenetic alterations have been implicated in brain plasticity and are highly responsive to experience, these alterations are promising candidates for molecular substrates of spinal plasticity as well. Thus, the current study investigated behavioral changes in the development of weight-bearing locomotion and epigenetic modifications in the spinal cord of infant rats following a neonatal low-thoracic spinal transection or sham surgery on postnatal day (P)1. Specifically, global levels of methylation and methylation status of the brain-derived neurotrophic factor (Bdnf) gene, a neurotrophin heavily involved in both CNS and behavioral plasticity, particularly in development, were examined in lumbar tissue harvested on P10 from sham and spinal-transected subjects. Behavioral results demonstrate that compared to shams, spinal-transected subjects exhibit significantly reduced partial-weight bearing hindlimb activity. Molecular data demonstrate group differences in global lumbar methylation levels as well as exon-specific group differences in Bdnf methylation. This study represents an initial step toward understanding the relationship between epigenetic mechanisms and plasticity associated with spinal cord and locomotor development.


Asunto(s)
Metilación de ADN/fisiología , Locomoción/fisiología , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/metabolismo , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epigénesis Genética/fisiología , Masculino , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/psicología
11.
Sci Rep ; 9(1): 10253, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31311968

RESUMEN

The quality of parental care received during development profoundly influences an individual's phenotype, including that of maternal behavior. We previously found that female rats with a history of maltreatment during infancy mistreat their own offspring. One proposed mechanism through which early-life experiences influence behavior is via epigenetic modifications. Indeed, our lab has identified a number of brain epigenetic alterations in female rats with a history of maltreatment. Here we sought to investigate the role of DNA methylation in aberrant maternal behavior. We administered zebularine, a drug known to alter DNA methylation, to dams exposed during infancy to the scarcity-adversity model of low nesting resources, and then characterized the quality of their care towards their offspring. First, we replicate that dams with a history of maltreatment mistreat their own offspring. Second, we show that maltreated-dams treated with zebularine exhibit lower levels of adverse care toward their offspring. Third, we show that administration of zebularine in control dams (history of nurturing care) enhances levels of adverse care. Lastly, we show altered methylation and gene expression in maltreated dams normalized by zebularine. These findings lend support to the hypothesis that epigenetic alterations resulting from maltreatment causally relate to behavioral outcomes.


Asunto(s)
Citidina/análogos & derivados , Metilación de ADN/efectos de los fármacos , Conducta Materna/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/genética , Citidina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Conducta Materna/fisiología , Conducta Materna/psicología , Área Preóptica/efectos de los fármacos , Ratas Long-Evans , Razón de Masculinidad
12.
Int J Dev Neurosci ; 78: 178-184, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31075305

RESUMEN

Reorganization of the brain's epigenetic landscape occurs alongside early adversity in both human and non-human animals. Whether this reorganization is simply incidental to or is a causal mechanism of the behavioral abnormalities that result from early adversity is important to understand. Using the scarcity-adversity model of low nesting resources in Long Evans rats, our lab has previously reported specific epigenetic and behavioral trajectories occurring in response to early disruption of the caregiving environment. To further probe that relationship, the current work investigates the ability of the epigenome-modifying drug sodium butyrate to prevent maltreatment-induced methylation changes when administered alongside maltreatment. Following exposure to the scarcity-adversity model, during which drug was administered prior to each caregiving session, methylation of Brain-derived Neurotrophic Factor (Bdnf) IX DNA was examined in the Prefrontal Cortex (PFC) of male and female pups at postnatal day (PN) 8. As our previous work reports, increased methylation at this exon of Bdnf in the PFC is a stable epigenetic change across the lifespan that occurs in response to early maltreatment, thus giving us a suitable starting point to investigate pharmacological prevention of maltreatment-induced epigenetic marks. Here we also examined off-target effects of sodium butyrate by assessing methylation in another region of Bdnf (exon IV) not affected in the infant brain as well as global levels of methylation in the brain region of interest. Results indicate that a 400 mg/kg (but not 300 mg/kg) dose of sodium butyrate is effective in preventing the maltreatment-induced rise in methylation at Bdnf exon IX in the PFC of male (but not female) infant pups. Administration of sodium butyrate did not affect the methylation status of Bdnf IV or overall levels of global methylation in the PFC, suggesting potential specificity of this drug. These data provide us an avenue forward for investigating whether the relationship between adversity-induced epigenetic outcomes in our model can be manipulated to improve behavioral outcomes.


Asunto(s)
Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epigénesis Genética , Conducta Materna , Corteza Prefrontal/metabolismo , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Ácido Butírico/farmacología , Metilación de ADN , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Masculino , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Long-Evans
13.
Dev Psychobiol ; 61(6): 824-831, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30810229

RESUMEN

The effects of exposure to developmental stress often diverge for males and females. Using the scarcity-adversity model of low nesting resources outside the home cage, our lab has discovered sex differences in both behavioral and epigenetic consequences of repeated exposure to caregiver maltreatment. For the measures we have performed to date, we have found more consequences for females. The reasons underlying this sex disparity are unknown. In the current experiment, we aimed to discern the quality of maternal care received by male and female pups in our model. As we have previously found more behavioral and epigenetic consequences in females, we hypothesized that females receive more adverse care compared to their male littermates. Our hypothesis was supported; in our maltreatment condition, we found that female pups received more adverse care than males. This sex difference in adverse care was not present in our two control conditions (cross-foster and normal maternal care). These data lend support to the notion that one reason females in our model incur more behavioral and epigenetic consequences is a result of greater mistreatment by the dam.


Asunto(s)
Conducta Animal/fisiología , Conducta Materna/fisiología , Caracteres Sexuales , Estrés Psicológico , Animales , Femenino , Masculino , Modelos Animales , Ratas , Ratas Long-Evans , Factores Sexuales
14.
Genes (Basel) ; 10(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650619

RESUMEN

The use of non-human animals in research is a longstanding practice to help us understand and improve human biology and health. Animal models allow researchers, for example, to carefully manipulate environmental factors in order to understand how they contribute to development, behavior, and health. In the field of behavioral epigenetics such approaches have contributed novel findings of how the environment physically interacts with our genes, leading to changes in behavior and health. This review highlights some of this research, focused on prenatal immune challenges, environmental toxicants, diet, and early-life stress. In conjunction, we also discuss why animal models were integral to these discoveries and the translational relevance of these discoveries.


Asunto(s)
Ambiente , Epigénesis Genética , Estrés Fisiológico/genética , Animales , Conducta Animal , Interacción Gen-Ambiente , Humanos , Estrés Fisiológico/inmunología
15.
Dev Psychobiol ; 61(3): 323-340, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30426484

RESUMEN

Advances in epigenetic methodologies have deepened theoretical explanations of mechanisms linking early life stress (ELS) and disease outcomes and suggest promising targets for intervention. To date, however, human studies have not capitalized on the richness of diverse animal models to derive and systematically evaluate specific and testable hypotheses. To promote cross-species dialog and scientific advance, here we provide a classification scheme to systematically evaluate the match between characteristics of human and animal studies of ELS and DNA methylation. Three preclinical models were selected that are highly cited, and that differ in the nature and severity of the ELS manipulation as well as in the affected epigenetic loci (the licking and grooming, maternal separation, and caregiver maltreatment models). We evaluated the degree to which human studies matched these preclinical models with respect to the timing of ELS and of DNA methylation assessment, as well as the type of ELS, whether sex differences were explicitly examined, the tissue sampled, and the targeted loci. Results revealed <50% match (range of 8-83%) between preclinical models and human work on these variables. Immediate and longer-term suggestions to improve translational specificity are offered, with the goal of accelerating scientific advance.


Asunto(s)
Metilación de ADN , Epigénesis Genética , Modelos Animales , Estrés Psicológico , Investigación Biomédica Traslacional , Animales , Femenino , Humanos , Masculino
16.
Front Behav Neurosci ; 12: 126, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30008666

RESUMEN

Exposure to adversity early in development alters brain and behavioral trajectories. Data continue to accumulate that epigenetic mechanisms are a mediating factor between early-life adversity and adult behavioral phenotypes. Previous work from our laboratory has shown that female Long-Evans rats exposed to maltreatment during infancy display both aberrant forced swim behavior and patterns of brain DNA methylation in adulthood. Therefore, we examined the possibility of rescuing the aberrant forced swim behavior in maltreated-adult females by administering an epigenome-modifying drug (zebularine) at a dose previously shown to normalize DNA methylation. We found that zebularine normalized behavior in the forced swim test in maltreated females such that they performed at the levels of controls (females that had been exposed to only nurturing care during infancy). These data help link DNA methylation to an adult phenotype in our maltreatment model, and more broadly provide additional evidence that non-targeted epigenetic manipulations can change behavior associated with early-life adversity.

17.
Prog Mol Biol Transl Sci ; 157: 1-19, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29933946

RESUMEN

It is understood that adversity during development has the power to alter behavioral trajectories, and the role of the epigenome in that relationship is currently under intense investigation. Several studies in both nonhuman animals and humans have established a link between early adversity and epigenetic regulation of genes heavily implicated in the stress response, plasticity and cognition, and psychiatric disorders such as depression and anxiety. Thus the relatively recent surge of studies centering on the epigenetic outcomes of stress has great potential to inform treatments and interventions for psychiatric disorder precipitated by early adversity. Here we review what we know and what we do not know, and suggest approaches to help further elucidate the relationship between early adversity, epigenetics, and behavior.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Epigénesis Genética , Estrés Psicológico/genética , Animales , Metilación de ADN/genética , Femenino , Humanos , Longevidad , Masculino , Caracteres Sexuales
18.
Policy Insights Behav Brain Sci ; 4(2): 202-209, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29202007

RESUMEN

Nature and nurture work together to drive development, behavior, and health. Behavioral epigenetics research has uncovered the underlying mechanisms for how this happens. Children's early years in development may offer the greatest opportunity for environmental and experiential factors to influence epigenome (chemical compounds telling our genes what to do), but evidence suggests it is never too late. The policy implications of this research are vast, including relevance for child development, health, and disease intervention and prevention.

19.
Dev Psychobiol ; 59(6): 703-714, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28767135

RESUMEN

Early life adversity is known to disrupt behavioral trajectories and many rodent models have been developed to characterize these stress-induced outcomes. One example is the scarcity-adversity model of low nesting resources. This model employs resource scarcity (i.e., low nesting materials) to elicit adverse caregiving conditions (including maltreatment) toward rodent neonates. Our lab utilizes a version of this model wherein caregiving exposures occur outside the home cage during the first postnatal week. The aim of this study was to determine adolescent and adult phenotypic outcomes associated with this model, including assessment of depressive- and anxiety-like behaviors and performance in different cognitive domains. Exposure to adverse caregiving had no effect on adolescent behavioral performance whereas exposure significantly impaired adult behavioral performance. Further, adult behavioral assays revealed substantial differences between sexes. Overall, data demonstrate the ability of repeated exposure to brief bouts of maltreatment outside the home cage in infancy to impact the development of several behavioral domains later in life.


Asunto(s)
Conducta Animal/fisiología , Conducta de Elección/fisiología , Conducta Materna/fisiología , Estrés Psicológico/fisiopatología , Animales , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Femenino , Vivienda para Animales , Masculino , Ratas , Ratas Long-Evans , Caracteres Sexuales
20.
Stress ; 20(5): 421-448, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28617197

RESUMEN

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.


Asunto(s)
Maltrato a los Niños , Cognición , Emociones , Conducta Materna , Comportamiento de Nidificación , Estrés Psicológico/psicología , Tejido Adiposo Blanco/metabolismo , Animales , Animales Recién Nacidos , Ropa de Cama y Ropa Blanca , Conducta Animal , Epigénesis Genética , Femenino , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Recién Nacido , Masculino , Modelos Animales , Neurogénesis , Sistema Hipófiso-Suprarrenal/metabolismo , Reproducibilidad de los Resultados , Resiliencia Psicológica , Roedores , Factores Sexuales , Estrés Psicológico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA