Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain Behav Immun ; 96: 239-255, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34126173

RESUMEN

Many neurodevelopmental disorders and associated learning deficits have been linked to early-life immune activation or ongoing immune dysregulation (Laskaris et al., 2016; O'Connor et al., 2014; Frick et al., 2013). Neuroscientists have begun to understand how the maturation of neural circuits allows for the emergence of cognitive and learning behaviors; yet we know very little about how these developing neural circuits are perturbed by certain events, including risk-factors such as early-life immune activation and immune dysregulation. To answer these questions, we examined the impact of early-life immune activation on the emergence of hippocampal-dependent learning in juvenile male and female rats using a well-characterized hippocampal-dependent learning task and we investigated the corresponding, dynamic multicellular interactions in the hippocampus that may contribute to these learning deficits. We found that even low levels of immune activation can result in hippocampal-depedent learning deficits days later, but only when this activation occurs during a sensitive period of development. The initial immune response and associated cytokine production in the hippocampus resolved within 24 h, several days prior to the observed learning deficit, but notably the initial immune response was followed by altered microglial-neuronal communication and synapse remodeling that changed the structure of hippocampal neurons during this period of juvenile brain development. We conclude that immune activation or dysregulation during a sensitive period of hippocampal development can precipitate the emergence of learning deficits via a multi-cellular process that may be initiated by, but not the direct result of the initial cytokine response. SIGNIFICANCE STATEMENT: Many neurodevelopmental disorders have been linked to early-life immune activation or immune dysregulation; however, very little is known about how dynamic changes in neuroimmune cells mediate the transition from normal brain function to the early stages of cognitive disorders, or how changes in immune signaling are subsequently integrated into developing neuronal networks. The current experiments examined the consequences of immune activation on the cellular and molecular changes that accompany the emergence of learning deficits during a sensitive period of hippocampal development. These findings have the potential to significantly advance our understanding of how early-life immune activation or dysregulation can result in the emergence of cognitive and learning deficits that are the largest source of years lived with disability in humans.


Asunto(s)
Hipocampo , Microglía , Animales , Femenino , Masculino , Plasticidad Neuronal , Neuronas , Ratas , Sinapsis
2.
J Immunol ; 202(7): 2131-2140, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30737275

RESUMEN

Puberty is a critical period of development marked by sexual, immune, and neural maturation. Exposure to stress during this period can lead to enduring changes in brain functioning and in behavior; however, the underlying mechanisms and the programming effects of stress during puberty remain unknown. Therefore, the objective of this study was to investigate the programming effects of pubertal immune challenge in response to a homotypic stressor later in life in CD-1 mice. Age and sex differences in the peripheral and central cytokine levels, along with sickness behavior and telemetry data, were analyzed following the secondary treatment. The results showed that pretreatment with LPS attenuated the immune response to a second homotypic challenge. Males pretreated with LPS during puberty and in early adulthood displayed an attenuated hypothermic response following the second LPS treatment compared with saline-pretreated controls, which is consistent with the attenuated peripheral IL-6 and IFN-γ concentrations. Females pretreated with LPS during puberty displayed lower IL-1ß, TNF-α, and IL-6 mRNA expression in the prefrontal cortex following the secondary immune challenge compared with saline controls. The results of this study show that exposure to LPS during puberty programs the peripheral and central immune responses, resulting in an attenuated immune response following a subsequent homotypic stressor. Thus, exposure to an immune challenge during puberty affects immune function later in life, which could permanently affect brain function and have implications on mental health.


Asunto(s)
Fenómenos del Sistema Inmunológico/efectos de los fármacos , Lipopolisacáridos/toxicidad , Maduración Sexual/inmunología , Estrés Fisiológico/inmunología , Animales , Femenino , Masculino , Ratones
3.
Front Neuroendocrinol ; 55: 100791, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31542287

RESUMEN

Adolescence marks a key developmental window during which emotion dysregulation increases, along with risk for the onset of anxiety and other affect-related pathologies. Although emotion dysregulation and related pathologies normatively decline during the transition into adulthood, this does not occur for a sizable minority of individuals. Finally, sex differences in anxiety emerge during adolescence, with females developing a 2-fold increase in risk relative to males. Unfortunately, a neurobiological model of the mechanisms that cause these changes during adolescence has yet to be proposed. In the present work, we first provide brief reviews of relevant literature. Next, we outline a dual-mechanism model focused on (i) the influence of pubertal testosterone on key emotion-regulation circuitry (i.e., orbitofrontal cortex-amygdala coupling) and (ii) myelination of the fiber bundles connecting such circuitry (i.e., uncinate fasciculus). The proposed model offers a set of specific, testable hypotheses that will hopefully spur much needed cross-disciplinary research.


Asunto(s)
Síntomas Afectivos , Amígdala del Cerebelo , Trastornos de Ansiedad , Red Nerviosa , Sistemas Neurosecretores/metabolismo , Corteza Prefrontal , Pubertad/metabolismo , Caracteres Sexuales , Testosterona/metabolismo , Adolescente , Adulto , Síntomas Afectivos/metabolismo , Síntomas Afectivos/fisiopatología , Amígdala del Cerebelo/crecimiento & desarrollo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiopatología , Animales , Trastornos de Ansiedad/metabolismo , Trastornos de Ansiedad/fisiopatología , Femenino , Humanos , Masculino , Red Nerviosa/crecimiento & desarrollo , Red Nerviosa/metabolismo , Red Nerviosa/fisiopatología , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Adulto Joven
4.
Front Neuroendocrinol ; 51: 25-35, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29110974

RESUMEN

Pregnancy is associated with a number of significant changes in maternal physiology. Perhaps one of the more notable changes is the significant alteration in immune function that occurs during pregnancy. This change in immune function is necessary to support a successful pregnancy, but also creates a unique period of life during which a female is susceptible to disease and, as we'll speculate here, may also contribute to mental health disorders associated with pregnancy and the postpartum period. Here, we review the known changes in peripheral immune function that occur during pregnancy and the postpartum period, while highlighting the impact of hormones during these times on immune function, brain or neural function, as well as behavior. We also discuss the known and possible impact of pregnancy-induced immune changes on neural function during this time and briefly discuss how these changes might be a risk factor for perinatal anxiety or mood disorders.


Asunto(s)
Trastornos de Ansiedad , Trastornos del Humor , Periodo Posparto , Embarazo , Psiconeuroinmunología , Trastornos Puerperales , Animales , Trastornos de Ansiedad/etiología , Trastornos de Ansiedad/inmunología , Trastornos de Ansiedad/metabolismo , Femenino , Humanos , Trastornos del Humor/etiología , Trastornos del Humor/inmunología , Trastornos del Humor/metabolismo , Periodo Posparto/inmunología , Periodo Posparto/metabolismo , Embarazo/inmunología , Embarazo/metabolismo , Trastornos Puerperales/inmunología , Trastornos Puerperales/metabolismo
5.
Horm Behav ; 114: 104521, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30981689

RESUMEN

Pregnancy and the postpartum period are periods of significant change in the immune and endocrine systems. This period of life is also associated with an increased risk of mental health disorders in the mother, and an increased risk of developmental and neuropsychiatric disorders in her infant. The collective data described here supports the idea that peripartum mood disorders in mother and developmental disorders in her infant likely reflects multiple pathogeneses, stemming from various interactions between the immune, endocrine and nervous systems, thereby resulting in various symptom constellations. In this case, testing the mechanisms underlying specific symptoms of these disorders (e.g. deficits in specific types of learning or anhedonia) may provide a better understanding of the various physiological interactions and multiple etiologies that most likely underlie the risk of mental health disorders during this unique time in life. The goal here is to summarize the current understanding of how immune and endocrine factors contribute to maternal mental health, while simultaneously understanding the impact these unique interactions have on the developing brain of her infant.


Asunto(s)
Investigación Biomédica/tendencias , Salud Mental/tendencias , Madres/psicología , Neuropsiquiatría/tendencias , Relaciones Padres-Hijo , Distinciones y Premios , Encéfalo/crecimiento & desarrollo , Sistema Endocrino/fisiología , Femenino , Humanos , Inmunidad Innata/fisiología , Lactante , Trastornos Mentales/etiología , Trastornos del Humor/complicaciones , Trastornos del Humor/fisiopatología , Trastornos del Humor/psicología , Fenómenos Fisiológicos del Sistema Nervioso , Periodo Posparto/psicología , Embarazo , Trastornos Puerperales/fisiopatología , Trastornos Puerperales/psicología , Salud de la Mujer
6.
Brain Behav Immun ; 66: 201-209, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28669797

RESUMEN

There is strong evidence that the immune system changes dramatically during pregnancy in order to prevent the developing fetus from being "attacked" by the maternal immune system. Due to these alterations in peripheral immune function, many women that suffer from autoimmune disorders actually find significant relief from their symptoms throughout pregnancy; however, these changes can also leave the mother more susceptible to infections that would otherwise be mitigated by the inflammatory response (Robinson and Klein, 2012). Only one other study has looked at changes in microglial number and morphology during pregnancy and the postpartum period (Haim et al., 2016), but no one has yet examined the neuroimmune response following an immune challenge during this time. Therefore, in this study, we investigated the impact of an immune challenge during various time-points throughout pregnancy and the postpartum period on the expression of immune molecules in the brain of the mother and fetus. Our results indicate that similar to the peripheral immune suppression measured during pregnancy, we also see significant suppression of the immune response in the maternal brain, particularly during late gestation. In contrast to the peripheral immune system, immune modulation in the maternal brain extends moderately into the postpartum period. Additionally, we found that the fetal immune response in the brain and placenta is also suppressed just before parturition, suggesting that cytokine production in the fetus and placenta are mirroring the peripheral cytokine response of the mother.


Asunto(s)
Encéfalo/inmunología , Feto/inmunología , Inflamación/inmunología , Periodo Posparto , Complicaciones del Embarazo/inmunología , Embarazo/inmunología , Animales , Femenino , Hipocampo/inmunología , Inflamación/complicaciones , Lipopolisacáridos/administración & dosificación , Neuroinmunomodulación , Placenta/inmunología , Corteza Prefrontal/inmunología , Área Preóptica/inmunología , Ratas Sprague-Dawley , Bazo/inmunología
7.
J Neurosci ; 33(3): 961-71, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325235

RESUMEN

Adolescence in humans represents a unique developmental time point associated with increased risk-taking behavior and experimentation with drugs of abuse. We hypothesized that exposure to drugs of abuse during adolescence may increase the risk of addiction in adulthood. To test this, rats were treated with a subchronic regimen of morphine or saline in adolescence, and their preference for morphine was examined using conditioned place preference (CPP) and drug-induced reinstatement in adulthood. The initial preference for morphine did not differ between groups; however, rats treated with morphine during adolescence showed robust reinstatement of morphine CPP after drug re-exposure in adulthood. This effect was not seen in rats pretreated with a subchronic regimen of morphine as adults, suggesting that exposure to morphine specifically during adolescence increases the risk of relapse to drug-seeking behavior in adulthood. We have previously established a role for microglia, the immune cells of the brain, and immune molecules in the risk of drug-induced reinstatement of morphine CPP. Thus, we examined the role of microglia within the nucleus accumbens of these rats and determined that rats exposed to morphine during adolescence had a significant increase in Toll-like receptor 4 (TLR4) mRNA and protein expression specifically on microglia. Morphine binds to TLR4 directly, and this increase in TLR4 was associated with exaggerated morphine-induced TLR4 signaling and microglial activation in rats previously exposed to morphine during adolescence. These data suggest that long-term changes in microglial function, caused by adolescent morphine exposure, alter the risk of drug-induced reinstatement in adulthood.


Asunto(s)
Microglía/efectos de los fármacos , Morfina/farmacología , Narcóticos/farmacología , Núcleo Accumbens/efectos de los fármacos , Trastornos Relacionados con Opioides/inmunología , Factores de Edad , Animales , Condicionamiento Operante/efectos de los fármacos , Modelos Animales de Enfermedad , Comportamiento de Búsqueda de Drogas , Microglía/metabolismo , Microglía/fisiología , Minociclina/farmacología , Núcleo Accumbens/inmunología , Núcleo Accumbens/metabolismo , Trastornos Relacionados con Opioides/metabolismo , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Recurrencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
8.
Cells ; 13(18)2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39329733

RESUMEN

Neonatal hypoxic-ischemic encephalopathy (HIE) occurs in 1.5 per 1000 live births, leaving affected children with long-term motor and cognitive deficits. Few animal models of HIE incorporate maternal immune activation (MIA) despite the significant risk MIA poses to HIE incidence and diagnosis. Our non-invasive model of HIE pairs late gestation MIA with postnatal hypoxia. HIE pups exhibited a trend toward smaller overall brain size and delays in the ontogeny of several developmental milestones. In adulthood, HIE animals had reduced strength and gait deficits, but no difference in speed. Surprisingly, HIE animals performed better on the rotarod, an assessment of motor coordination. There was significant upregulation of inflammatory genes in microglia 24 h after hypoxia. Single-cell RNA sequencing (scRNAseq) revealed two microglia subclusters of interest following HIE. Pseudobulk analysis revealed increased microglia motility gene expression and upregulation of epigenetic machinery and neurodevelopmental genes in macrophages following HIE. No sex differences were found in any measures. These results support a two-hit noninvasive model pairing MIA and hypoxia as a model for HIE in humans. This model results in a milder phenotype compared to established HIE models; however, HIE is a clinically heterogeneous injury resulting in a variety of outcomes in humans. The pathways identified in our model of HIE may reveal novel targets for therapy for neonates with HIE.


Asunto(s)
Animales Recién Nacidos , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica , Inflamación , Microglía , Monocitos , Animales , Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/metabolismo , Ratones , Inflamación/patología , Inflamación/genética , Monocitos/metabolismo , Femenino , Microglía/metabolismo , Microglía/patología , Masculino , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Ratones Endogámicos C57BL , Trastornos Motores/genética , Trastornos Motores/patología
9.
Front Neuroendocrinol ; 33(3): 267-86, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22982535

RESUMEN

The brain, endocrine, and immune systems are inextricably linked. Immune molecules have a powerful impact on neuroendocrine function, including hormone-behavior interactions, during health as well as sickness. Similarly, alterations in hormones, such as during stress, can powerfully impact immune function or reactivity. These functional shifts are evolved, adaptive responses that organize changes in behavior and mobilize immune resources, but can also lead to pathology or exacerbate disease if prolonged or exaggerated. The developing brain in particular is exquisitely sensitive to both endogenous and exogenous signals, and increasing evidence suggests the immune system has a critical role in brain development and associated behavioral outcomes for the life of the individual. Indeed, there are associations between many neuropsychiatric disorders and immune dysfunction, with a distinct etiology in neurodevelopment. The goal of this review is to describe the important role of the immune system during brain development, and to discuss some of the many ways in which immune activation during early brain development can affect the later-life outcomes of neural function, immune function, mood and cognition.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Sistema Inmunológico/crecimiento & desarrollo , Afecto , Animales , Animales Recién Nacidos , Astrocitos/fisiología , Conducta , Encéfalo/inmunología , Trastornos del Conocimiento/etiología , Citocinas/fisiología , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/fisiopatología , Inmunidad Innata/fisiología , Interleucina-1beta/fisiología , Lipopolisacáridos/toxicidad , Trastornos Mentales/fisiopatología , Microglía/inmunología , Microglía/fisiología , Modelos Animales , Neurogénesis/fisiología , Sistemas Neurosecretores/fisiología , Poli I-C/toxicidad
10.
Front Neurosci ; 17: 1135559, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37123361

RESUMEN

Epidemiological evidence suggests that one's risk of being diagnosed with a neurodevelopmental disorder (NDD)-such as autism, ADHD, or schizophrenia-increases significantly if their mother had a viral or bacterial infection during the first or second trimester of pregnancy. Despite this well-known data, little is known about how developing neural systems are perturbed by events such as early-life immune activation. One theory is that the maternal immune response disrupts neural processes important for typical fetal and postnatal development, which can subsequently result in specific and overlapping behavioral phenotypes in offspring, characteristic of NDDs. As such, rodent models of maternal immune activation (MIA) have been useful in elucidating neural mechanisms that may become dysregulated by MIA. This review will start with an up-to-date and in-depth, critical summary of epidemiological data in humans, examining the association between different types of MIA and NDD outcomes in offspring. Thereafter, we will summarize common rodent models of MIA and discuss their relevance to the human epidemiological data. Finally, we will highlight other factors that may interact with or impact MIA and its associated risk for NDDs, and emphasize the importance for researchers to consider these when designing future human and rodent studies. These points to consider include: the sex of the offspring, the developmental timing of the immune challenge, and other factors that may contribute to individual variability in neural and behavioral responses to MIA, such as genetics, parental age, the gut microbiome, prenatal stress, and placental buffering.

11.
J Neuroendocrinol ; 35(7): e13275, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37186019

RESUMEN

This study examined the effect of limited bedding and nesting (LBN) stress on postpartum anhedonia, maternal behaviors, anxiety-like behaviors, and neuroendocrine and neuroimmune function as a potential model of postpartum depression. Dams underwent sucrose preference tests prior to breeding, during gestation and again postpartum, to examine the potential onset of anhedonia. On embryonic day 19, dams were placed into either a LBN or control housing condition. Contrary to our predictions, LBN stress had no effect on postpartum sucrose preference. We also found no effect of LBN condition on fecal estradiol or corticosterone levels, both of which increased at birth and decreased postpartum. Regardless of housing conditions, approximately 40% of new mothers exhibited a decrease in sucrose preference, while others show no change, suggesting an individual susceptibility to postpartum anhedonia. In a separate cohort of LBN and control dams, we measured pup retrieval, hoarding behavior, elevated plus maze (EPM), and marble burying. LBN dams exhibited increased anxiety, associated with decreased time spent in the open arms of the EPM. We also measured a significant increase in IL-6 expression in the dorsal hippocampus and medial prefrontal cortex of postpartum dams compared to nonpregnant dams. These findings suggest that while LBN stress has effects on anxiety and maternal care, it does not induce postpartum anhedonia. Rather, there are inherent differences in susceptibility to anhedonia in individual dams, and future studies should be conducted to better understand individual vulnerability and resilience to postpartum anhedonia.


Asunto(s)
Anhedonia , Depresión Posparto , Femenino , Humanos , Ratas , Animales , Ratas Sprague-Dawley , Periodo Posparto , Sacarosa/farmacología , Estrés Psicológico
12.
J Neurosci ; 31(49): 17835-47, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22159099

RESUMEN

A critical component of drug addiction research involves identifying novel biological mechanisms and environmental predictors of risk or resilience to drug addiction and associated relapse. Increasing evidence suggests microglia and astrocytes can profoundly affect the physiological and addictive properties of drugs of abuse, including morphine. We report that glia within the rat nucleus accumbens (NAcc) respond to morphine with an increase in cytokine/chemokine expression, which predicts future reinstatement of morphine conditioned place preference (CPP) following a priming dose of morphine. This glial response to morphine is influenced by early-life experience. A neonatal handling paradigm that increases the quantity and quality of maternal care significantly increases baseline expression of the anti-inflammatory cytokine IL-10 within the NAcc, attenuates morphine-induced glial activation, and prevents the subsequent reinstatement of morphine CPP in adulthood. IL-10 expression within the NAcc and reinstatement of CPP are negatively correlated, suggesting a protective role for this specific cytokine against morphine-induced glial reactivity and drug-induced reinstatement of morphine CPP. Neonatal handling programs the expression of IL-10 within the NAcc early in development, and this is maintained into adulthood via decreased methylation of the IL-10 gene specifically within microglia. The effect of neonatal handling is mimicked by pharmacological modulation of glia in adulthood with ibudilast, which increases IL-10 expression, inhibits morphine-induced glial activation within the NAcc, and prevents reinstatement of morphine CPP. Taken together, we have identified a novel gene × early-life environment interaction on morphine-induced glial activation and a specific role for glial activation in drug-induced reinstatement of drug-seeking behavior.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Interleucina-10/sangre , Acontecimientos que Cambian la Vida , Microglía/metabolismo , Morfina/farmacología , Narcóticos/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Cromatografía Liquida , Corticosterona/sangre , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/fisiología , Epigenómica , Extinción Psicológica/efectos de los fármacos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Manejo Psicológico , Inmunoprecipitación , Interleucina-10/genética , Masculino , Espectrometría de Masas , Microglía/efectos de los fármacos , Microinyecciones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología
13.
J Neurochem ; 120(6): 948-63, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22182318

RESUMEN

Microglia are the resident immune cells within the brain and their production of immune molecules such as cytokines and chemokines is critical for the processes of normal brain development including neurogenesis, axonal migration, synapse formation, and programmed cell death. Notably, sex differences exist in many of these processes throughout brain development; however, it is unknown whether a sex difference concurrently exists in the colonization, number, or morphology of microglia within the developing brain. We demonstrate for the first time that the number and morphology of microglia throughout development is dependent upon the sex and age of the individual, as well as the brain region of interest. Males have overall more microglia early in postnatal development [postnatal day (P) 4], whereas females have more microglia with an activated/amoeboid morphology later in development, as juveniles and adults (P30-60). Finally, gene expression of a large number of cytokines, chemokines and their receptors shifts dramatically over development, and is highly dependent upon sex. Taken together, these data warrant further research into the role that sex-dependent mechanisms may play in microglial colonization, number, and function, and their potential contribution to neural development, function, or potential dysfunction.


Asunto(s)
Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/fisiología , Microglía/fisiología , Caracteres Sexuales , Factores de Edad , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Quimiocina CXCL9/metabolismo , Embrión de Mamíferos , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
14.
Horm Behav ; 62(3): 243-53, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22387107

RESUMEN

Microglia and astrocytes are the primary immune cells within the central nervous system. Microglia influence processes including neural development, synaptic plasticity and cognition; while their activation and production of immune molecules can induce stereotyped sickness behaviors or pathologies including cognitive dysfunction. Given their role in health and disease, we propose that glia may also be a critical link in understanding the etiology of many neuropsychiatric disorders that present with a strong sex-bias in their symptoms or prevalence. Specifically, males are more likely to be diagnosed with disorders that have distinct developmental origins such as autism or schizophrenia. In contrast, females are more likely to be diagnosed with disorders that present later in life, after the onset of adolescence, such as depression and anxiety disorders. In this review we will summarize the evidence suggesting that sex differences in the colonization and function of glia within the normal developing brain may contribute to distinct windows of vulnerability between males and females. We will also highlight the current gaps in our knowledge as well as the future directions and considerations of research aimed at understanding the link between neuroimmune function and sex differences in mental health disorders.


Asunto(s)
Encéfalo/inmunología , Trastornos Mentales/inmunología , Neuroglía/inmunología , Caracteres Sexuales , Animales , Encéfalo/crecimiento & desarrollo , Femenino , Masculino
15.
J Neurosci Methods ; 374: 109567, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35306037

RESUMEN

BACKGROUND: To understand the role of microglia in brain function and development, methods have emerged to deplete microglia throughout the brain. Liposome-encapsulated clodronate (LEC) can be infused into the brain to deplete microglia in a brain-region and time-specific manner. NEW METHOD: This study validates methodology to deplete microglia in the rat dorsal hippocampus (dHP) during a specific period of juvenile development. Stereotaxic surgery was performed to infuse LEC at postnatal day (P) 16 or 19 into dHP. Rat brains were harvested at various ages to determine specificity of infusion and duration of depletion. RESULTS: P19 infusion of LEC into dHP with a 27G syringe depleted microglia in dHP subregions CA1, dentate gyrus (DG), and CA3 from P24-P30. There was also evidence of depletion in parietal cortex above the infusion site. P16 infusion of LEC with a 32 G syringe depleted microglia only in dHP subregions CA1 and DG from P21-P40. COMPARISON WITH EXISTING METHOD(S): Previous methods have infused LEC intra-hippocampally in adult rats or intra-cerebroventricularly in neonatal rats. This study is the first to publish methodology to deplete microglia in a brain-region specific manner during juvenile rat development. CONCLUSIONS: The timing of LEC infusion during the juvenile period can be adjusted to achieve maximal microglia depletion by a specific postnatal day. A 27G needle results in LEC backflow during the infusion, but also allows LEC to reach all subregions of dHP. Infusion with a 32 G needle prevents backflow during infusion, but results in a more local spread of LEC within dHP.


Asunto(s)
Hipocampo , Microglía , Animales , Encéfalo , Ácido Clodrónico , Ratas
16.
Horm Behav ; 59(3): 338-44, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20800064

RESUMEN

The establishment of sex-specific neural morphology, which underlies sex-specific behaviors, occurs during a perinatal sensitive window in which brief exposure to gonadal steroid hormones produces permanent masculinization of the brain. In the rodent, estradiol derived from testicular androgens is a principal organizational hormone. The mechanism by which transient estradiol exposure induces permanent differences in neuronal anatomy has been widely investigated, but remains elusive. Epigenetic changes, such as DNA methylation, allow environmental influences to alter long-term gene expression patterns and therefore may be a potential mediator of estradiol-induced organization of the neonatal brain. Here we review data that demonstrate sex and estradiol-induced differences in DNA methylation on the estrogen receptor α (ERα), estrogen receptor ß (ERß), and progesterone receptor (PR) promoters in sexually dimorphic brain regions across development. Contrary to the overarching view of DNA methylation as a permanent modification directly tied to gene expression, these data demonstrate that methylation patterns on steroid hormone receptors change across the life span and do not necessarily predict expression. Although further exploration into the mechanism and significance of estradiol-induced alterations in DNA methylation patterns in the neonatal brain is necessary, these results provide preliminary evidence that epigenetic alterations can occur in response to early hormone exposure and may mediate estradiol-induced organization of sex differences in the neonatal brain.


Asunto(s)
Encéfalo/metabolismo , Epigénesis Genética/genética , Estradiol/metabolismo , Receptores de Estrógenos/genética , Receptores de Progesterona/genética , Diferenciación Sexual/genética , Animales , Metilación de ADN/genética , Femenino , Masculino , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Caracteres Sexuales
17.
Behav Brain Res ; 415: 113449, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34252501

RESUMEN

Many individuals diagnosed with neuropsychiatric disorders, such as autism, attention-deficit/hyperactivity disorder, schizophrenia, and social anxiety disorder, all share a common dimension of aberrant social behavior. Epidemiological data indicate that adverse environmental factors contribute to the risk for neurodevelopmental disorders, including those associated with aberrant social behavior. Early-life exposure to infectious pathogens is one of those adverse environmental factors, suggesting that activation of the immune system during early development may contribute to disease pathology associated with altered social behavior. In the current project, we examined the impact of neonatal infection, with or without juvenile immune activation, on the expression of juvenile social behavior and on the expression of inflammatory cytokines and microglial signaling molecules in the juvenile rat brain. The outcomes of these experiments revealed that neonatal infection significantly decreased juvenile social interaction, but significantly increased juvenile play behavior in male and female rats. Moreover, neonatal infection alone, juvenile immune activation alone, and neonatal infection plus juvenile immune activation all significantly impaired social recognition in juvenile male rats. Juvenile female rats (including controls) did not demonstrate social recognition as measured in our three-chamber social recognition test. Taken together, the behavioral and molecular data presented here support the sensitivity of the developing brain to immune activation, particularly in the expression of age-appropriate social behaviors. These data warrant the design of additional studies to examine the mechanistic relationship between early-life immune activation and aberrant social behavior to develop novel as well as modify existing therapeutic targets and preventative measures to help those who display aberrant social behavior.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/inmunología , Citocinas/inmunología , Microglía/inmunología , Enfermedades Neuroinflamatorias/inmunología , Conducta Social , Cognición Social , Factores de Edad , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/inmunología , Femenino , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/efectos adversos , Masculino , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales
18.
Front Glob Womens Health ; 2: 726422, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34816242

RESUMEN

Purpose: The purpose of this study was to assess the association between various risk factors with postpartum depression severity using a large dataset that included variables such as previous mental health status, social factors, societal factors, health care access, and other state-wide or region-specific variables. Methods: We obtained the most recently available (2016-2017) dataset from the Pregnancy Risk Assessment Monitoring System (PRAMS), which is a dataset compiled by the Centers for Disease Control (CDC) that collects state-specific, population-based data on maternal attitudes and experiences before, during, and shortly after pregnancy from over 73,000 women in 39 states. We utilized a hierarchical linear model to analyze the data across various levels, with a symptom severity scale (0-8) as the dependent variable. Results: Of the 21 variables included in the final model, nine variables were statistically significant predictors of symptom severity. Statistically significant predictors of increased postpartum depression symptom severity included previous depression diagnosis and depression symptoms during pregnancy, baby not residing with mother, unintentional pregnancy, women with less than a high school degree and more than a college degree, Women Infants Children (WIC) enrollment, and married women. In contrast to these other factors, attendance at a postpartum follow up appointment was associated with significantly increased symptom severity. Age revealed an inverted curve in predicting postpartum symptom severity. Conclusions: There was no significant difference in symptom severity scores across the 39 participating states. Most notably, postpartum depression symptom severity was associated with previous depression diagnosis and previous symptom severity, but our results also reveal novel social and education factors that contribute to the support and well-being of the mother and child.

19.
Viruses ; 13(11)2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34835104

RESUMEN

Zika virus (ZIKV) is a mosquito-borne flavivirus that became widely recognized due to the epidemic in Brazil in 2015. Since then, there has been nearly a 20-fold increase in the incidence of microcephaly and birth defects seen among women giving birth in Brazil, leading the Centers for Disease Control and Prevention (CDC) to officially declare a causal link between prenatal ZIKV infection and the serious brain abnormalities seen in affected infants. Here, we used a unique rat model of prenatal ZIKV infection to study three possible long-term outcomes of congenital ZIKV infection: (1) behavior, (2) cell proliferation, survival, and differentiation in the brain, and (3) immune responses later in life. Adult offspring that were prenatally infected with ZIKV exhibited motor deficits in a sex-specific manner, and failed to mount a normal interferon response to a viral immune challenge later in life. Despite undetectable levels of ZIKV in the brain and serum in these offspring at P2, P24, or P60, these results suggest that prenatal exposure to ZIKV results in lasting consequences that could significantly impact the health of the offspring. To help individuals already exposed to ZIKV, as well as be prepared for future outbreaks, we need to understand the full spectrum of neurological and immunological consequences that could arise following prenatal ZIKV infection.


Asunto(s)
Exposición Materna/efectos adversos , Malformaciones del Sistema Nervioso/etiología , Trastornos del Neurodesarrollo/etiología , Complicaciones Infecciosas del Embarazo/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Infección por el Virus Zika , Animales , Animales Recién Nacidos , Femenino , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/fisiopatología
20.
Viruses ; 13(6)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34207958

RESUMEN

Since Zika virus (ZIKV) first emerged as a public health concern in 2015, our ability to identify and track the long-term neurological sequelae of prenatal Zika virus (ZIKV) infection in humans has been limited. Our lab has developed a rat model of maternal ZIKV infection with associated vertical transmission to the fetus that results in significant brain malformations in the neonatal offspring. Here, we use this model in conjunction with longitudinal magnetic resonance imaging (MRI) to expand our understanding of the long-term neurological consequences of prenatal ZIKV infection in order to identify characteristic neurodevelopmental changes and track them across time. We exploited both manual and automated atlas-based segmentation of MR images in order to identify long-term structural changes within the developing rat brain following inoculation. The paradigm involved scanning three cohorts of male and female rats that were prenatally inoculated with 107 PFU ZIKV, 107 UV-inactivated ZIKV (iZIKV), or diluent medium (mock), at 4 different postnatal day (P) age points: P2, P16, P24, and P60. Analysis of tracked brain structures revealed significantly altered development in both the ZIKV and iZIKV rats. Moreover, we demonstrate that prenatal ZIKV infection alters the growth of brain regions throughout the neonatal and juvenile ages. Our findings also suggest that maternal immune activation caused by inactive viral proteins may play a role in altered brain growth throughout development. For the very first time, we introduce manual and automated atlas-based segmentation of neonatal and juvenile rat brains longitudinally. Experimental results demonstrate the effectiveness of our novel approach for detecting significant changes in neurodevelopment in models of early-life infections.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Imagen por Resonancia Magnética/métodos , Trastornos del Neurodesarrollo/virología , Neuroimagen/métodos , Complicaciones Infecciosas del Embarazo/virología , Infección por el Virus Zika/complicaciones , Virus Zika/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Trastornos del Neurodesarrollo/diagnóstico por imagen , Embarazo , Complicaciones Infecciosas del Embarazo/diagnóstico por imagen , Ratas , Virus Zika/patogenicidad , Infección por el Virus Zika/diagnóstico por imagen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA