Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Horm Behav ; 158: 105470, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38061232

RESUMO

Hormonal contraceptives, including oral contraceptives (OCs), regulate hormonal cycles and broadly affect physiological processes, including stress responsivity. Whereas many users describe overall improved mood, up to 10 % of OC users experience adverse effects, including depression and anxiety. Given the link between regulation of hypothalamic-pituitary-adrenal (HPA) axis, stress exposure, and risk for depression, it is likely that OC-effects on stress mediate increased risk or increased resilience to these disorders. In this study, we developed and characterized a tractable mouse model of OC exposure with which to identify the mechanisms underlying OC modulation of brain, behavior, and mood. Specifically, we aimed to determine whether translationally relevant doses of OC-hormones in mice mimic changes in stress responsivity observed in humans taking OCs and describe behavioral changes during OC exposure. Young adult female C57Bl/6 N mice received daily ethinyl estradiol (EE) and levonorgestrel (LVNG) in 10 % sucrose, EE and drospirenone (DRSP) in 10 % sucrose, or 10 % sucrose alone. Translationally relevant doses of EE + LVNG-exposure, but not EE + DRSP, suppressed the acute stress response, consistent with effects observed in human OC users. EE + LVNG caused a specific anhedonia-like effect, without broad changes in stress-coping behavior, other depression-like behaviors, or anxiety-like behaviors. The suppression of regular estrous cycling, together with the blunting of the corticosterone response to acute stress, demonstrate the utility of this model for future studies to identify the mechanisms underlying OC interactions with stress, motivation, and risk for depression.


Assuntos
Anticoncepcionais Orais Combinados , Motivação , Humanos , Feminino , Animais , Camundongos , Depressão , Etinilestradiol/farmacologia , Sacarose
2.
Cells ; 12(4)2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36831303

RESUMO

Chromatin dysregulation has emerged as a major hallmark of neurodevelopmental disorders such as intellectual disability (ID) and autism spectrum disorders (ASD). The prevalence of ID and ASD is higher in males compared to females, with unknown mechanisms. Intellectual developmental disorder, X-linked syndromic, Claes-Jensen type (MRXSCJ), is caused by loss-of-function mutations of lysine demethylase 5C (KDM5C), a histone H3K4 demethylase gene. KDM5C escapes X-inactivation, thereby presenting at a higher level in females. Initially, MRXSCJ was exclusively reported in males, while it is increasingly evident that females with heterozygous KDM5C mutations can show cognitive deficits. The mouse model of MRXSCJ, male Kdm5c-hemizygous knockout animals, recapitulates key features of human male patients. However, the behavioral and molecular traits of Kdm5c-heterozygous female mice remain incompletely characterized. Here, we report that gene expression and behavioral abnormalities are readily detectable in Kdm5c-heterozygous female mice, demonstrating the requirement for a higher KDM5C dose in females. Furthermore, we found both shared and sex-specific consequences of a reduced KDM5C dose in social behavior, gene expression, and genetic interaction with the counteracting enzyme KMT2A. These observations provide an essential insight into the sex-biased manifestation of neurodevelopmental disorders and sex chromosome evolution.


Assuntos
Deficiência Intelectual , Transcriptoma , Humanos , Masculino , Feminino , Animais , Camundongos , Histona Desmetilases/metabolismo , Mutação , Cromatina , Deficiência Intelectual/genética
4.
eNeuro ; 10(3)2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36810148

RESUMO

Sign-tracking (ST) describes the propensity to approach and contact a Pavlovian reward cue. By contrast, goal-trackers (GTs) respond to such a cue by retrieving the reward. These behaviors index the presence of opponent cognitive-motivational traits, with STs exhibiting attentional control deficits, behavior dominated by incentive motivational processes, and vulnerability for addictive drug taking. Attentional control deficits in STs were previously attributed to attenuated cholinergic signaling, resulting from deficient translocation of intracellular choline transporters (CHTs) into synaptosomal plasma membrane. Here, we investigated a posttranslational modification of CHTs, poly-ubiquitination, and tested the hypothesis that elevated cytokine signaling in STs contributes to CHT modification. We demonstrated that intracellular CHTs, but not plasma membrane CHTs, are highly ubiquitinated in male and female sign-tracking rats when compared with GTs. Moreover, levels of cytokines measured in cortex and striatum, but not spleen, were higher in STs than in GTs. Activation of the innate immune system by systemic administration of the bacterial endotoxin lipopolysaccharide (LPS) elevated ubiquitinated CHT levels in cortex and striatum of GTs only, suggesting ceiling effects in STs. In spleen, LPS increased levels of most cytokines in both phenotypes. In cortex, LPS particularly robustly increased levels of the chemokines CCL2 and CXCL10. Phenotype-specific increases were restricted to GTs, again suggesting ceiling effects in STs. These results indicate that interactions between elevated brain immune modulator signaling and CHT regulation are essential components of the neuronal underpinnings of the addiction vulnerability trait indexed by sign-tracking.


Assuntos
Sinais (Psicologia) , Lipopolissacarídeos , Ratos , Masculino , Feminino , Animais , Ratos Sprague-Dawley , Lipopolissacarídeos/farmacologia , Motivação , Colinérgicos/farmacologia , Fenótipo , Recompensa
5.
Front Neuroendocrinol ; 67: 101035, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36075276

RESUMO

Hormonal contraceptives are among the most important health and economic developments in the 20thCentury, providing unprecedented reproductive control and a range of health benefits including decreased premenstrual symptoms and protections against various cancers. Hormonal contraceptives modulate neural function and stress responsivity. These changes are usually innocuous or even beneficial, including their effects onmood. However, in approximately 4-10% of users, or up to 30 million people at any given time, hormonal contraceptives trigger depression or anxiety symptoms. How hormonal contraceptives contribute to these responses and who is at risk for adverse outcomes remain unknown. In this paper, we discussstudies of hormonal contraceptive use in humans and describe the ways in which laboratory animal models of contraceptive hormone exposure will be an essential tool for expanding findings to understand the precise mechanisms by which hormonal contraceptives influence the brain, stress responses, and depression risk.


Assuntos
Encéfalo , Anticoncepcionais Orais Hormonais , Humanos , Feminino , Animais , Anticoncepcionais Orais Hormonais/efeitos adversos , Modelos Animais
7.
Neuron ; 109(20): 3206-3210, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34551313

RESUMO

Laboratories are the central workplace for academic scientists and can play a key role in supporting psychological safety, mental health, and well-being. We provide strategies to build inclusive structures within laboratories and support mental health for all members.


Assuntos
Pesquisa Biomédica , Educação de Pós-Graduação/organização & administração , Eficiência , Docentes/psicologia , Saúde Mental , Estudantes/psicologia , Local de Trabalho/psicologia , COVID-19 , Humanos , Laboratórios , Tutoria , Neurociências/educação , Política Organizacional , SARS-CoV-2
8.
J Neuroinflammation ; 18(1): 193, 2021 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-34488804

RESUMO

BACKGROUND: The neuroimmune system is required for normal neural processes, including modulation of cognition, emotion, and adaptive behaviors. Aberrant neuroimmune activation is associated with dysregulation of memory and emotion, though the precise mechanisms at play are complex and highly context dependent. Sex differences in neuroimmune activation and function further complicate our understanding of its roles in cognitive and affective regulation. METHODS: Here, we characterized the physiological sickness and inflammatory response of the hippocampus following intracerebroventricular (ICV) administration of a synthetic viral mimic, polyinosinic:polycytidylic acid (poly I:C), in both male and female C57Bl/6N mice. RESULTS: We observed that poly I:C induced weight loss, fever, and elevations of cytokine and chemokines in the hippocampus of both sexes. Specifically, we found transient increases in gene expression and protein levels of IL-1α, IL-1ß, IL-4, IL-6, TNFα, CCL2, and CXCL10, where males showed a greater magnitude of response compared with females. Only males showed increased IFNα and IFNγ in response to poly I:C, whereas both males and females exhibited elevations of IFNß, demonstrating a specific sex difference in the anti-viral response in the hippocampus. CONCLUSION: Our data suggest that type I interferons are one potential node mediating sex-specific cytokine responses and neuroimmune effects on cognition. Together, these findings highlight the importance of using both males and females and analyzing a broad set of inflammatory markers in order to identify the precise, sex-specific roles for neuroimmune dysregulation in neurological diseases and disorders.


Assuntos
Poli I-C , Caracteres Sexuais , Animais , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Poli I-C/farmacologia
9.
Commun Biol ; 3(1): 278, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32483278

RESUMO

Histone H3 lysine 4 methylation (H3K4me) is extensively regulated by numerous writer and eraser enzymes in mammals. Nine H3K4me enzymes are associated with neurodevelopmental disorders to date, indicating their important roles in the brain. However, interplay among H3K4me enzymes during brain development remains largely unknown. Here, we show functional interactions of a writer-eraser duo, KMT2A and KDM5C, which are responsible for Wiedemann-Steiner Syndrome (WDSTS), and mental retardation X-linked syndromic Claes-Jensen type (MRXSCJ), respectively. Despite opposite enzymatic activities, the two mouse models deficient for either Kmt2a or Kdm5c shared reduced dendritic spines and increased aggression. Double mutation of Kmt2a and Kdm5c clearly reversed dendritic morphology, key behavioral traits including aggression, and partially corrected altered transcriptomes and H3K4me landscapes. Thus, our study uncovers common yet mutually suppressive aspects of the WDSTS and MRXSCJ models and provides a proof of principle for balancing a single writer-eraser pair to ameliorate their associated disorders.


Assuntos
Anormalidades Múltiplas/genética , Agressão , Anormalidades Craniofaciais/genética , Espinhas Dendríticas/metabolismo , Transtornos do Crescimento/genética , Histona Desmetilases/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/metabolismo , Hipertricose/genética , Deficiência Intelectual/genética , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína de Leucina Linfoide-Mieloide/genética , Animais , Modelos Animais de Doenças , Histona Desmetilases/deficiência , Histona-Lisina N-Metiltransferase/deficiência , Masculino , Metilação , Camundongos , Proteína de Leucina Linfoide-Mieloide/deficiência
11.
Neuroscience ; 428: 76-89, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31917350

RESUMO

Major illnesses, including heart attack and sepsis, can cause cognitive impairments, depression, and progressive memory decline that persist long after recovery from the original illness. In rodent models of sepsis or subchronic immune challenge, memory deficits also persist for weeks or months, even in the absence of ongoing neuroimmune activation. This raises the question of what mechanisms in the brain mediate such persistent changes in neural function. Here, we used RNA-sequencing as a large-scale, unbiased approach to identify changes in hippocampal gene expression long after a subchronic immune challenge previously established to cause persistent memory impairments in both males and females. We observed enduring dysregulation of gene expression three months after the end of a subchronic immune challenge. Surprisingly, there were striking sex differences in both the magnitude of changes and the specific genes and pathways altered, where males showed persistent changes in both immune- and plasticity-related genes three months after immune challenge, whereas females showed few such changes. In contrast, females showed striking differential gene expression in response to a subsequent immune challenge. Thus, immune activation has enduring and sex-specific consequences for hippocampal gene expression and the transcriptional response to subsequent stimuli. Together with findings of long-lasting memory impairments after immune challenge, these data suggest that illnesses can cause enduring vulnerability to, cognitive decline, affective disorders, and memory impairments via dysregulation of transcriptional processes in the brain.


Assuntos
Encéfalo/fisiopatologia , Hipocampo/fisiopatologia , Transtornos da Memória/fisiopatologia , Memória/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica/fisiologia , Hipocampo/metabolismo , Masculino , Transtornos da Memória/metabolismo , Camundongos Endogâmicos C57BL , Caracteres Sexuais
13.
Trends Neurosci ; 42(10): 680-692, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31473031

RESUMO

Emerging research demonstrates that a pattern of overlapping but distinct molecular and circuit mechanisms are engaged by males and females during memory tasks. Importantly, sex differences in neural mechanisms and behavioral strategies are evident even when performance on a memory task is similar between females and males. We propose that sex differences in memory may be best understood within a dynamic memory systems framework. Specifically, sex differences in hormonal influences and neural circuit development result in biases in the circuits engaged and the information preferentially stored or retrieved in males and females. By using animal models to understand the neural networks and molecular mechanisms required for memory in both sexes, we can gain crucial insights into sex and gender biases in disorders including post-traumatic stress disorder (PTSD) in humans.


Assuntos
Encéfalo/fisiologia , Medo/psicologia , Memória/fisiologia , Rede Nervosa/fisiologia , Animais , Modelos Animais de Doenças , Medo/fisiologia , Feminino , Humanos , Masculino , Caracteres Sexuais , Transtornos de Estresse Pós-Traumáticos/fisiopatologia , Transtornos de Estresse Pós-Traumáticos/psicologia
14.
Behav Neurosci ; 133(4): 448-459, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30896190

RESUMO

Previous research emphasized the impact of traumatic brain injury on cholinergic systems and associated cognitive functions. Here we addressed the converse question: Because of the available evidence indicating cognitive and neuronal vulnerabilities in humans expressing low-capacity cholinergic systems or with declining cholinergic systems, do injuries cause more severe cognitive decline in such subjects, and what cholinergic mechanisms contribute to such vulnerability? Using mice heterozygous for the choline transporter (CHT+/- mice) as a model for a limited cholinergic capacity, we investigated the cognitive and neuronal consequences of repeated, mild concussion injuries (rmCc). After five rmCc, and compared with wild type (WT) mice, CHT+/- mice exhibited severe and lasting impairments in sustained attention performance, consistent with effects of cholinergic losses on attention. However, rmCc did not affect the integrity of neuronal cell bodies and did not alter the density of cortical synapses. As a cellular mechanism potentially responsible for the attentional impairment in CHT+/- mice, we found that rmCc nearly completely attenuated performance-associated, CHT-mediated choline transport. These results predict that subjects with an already vulnerable cholinergic system will experience severe and lasting cognitive-cholinergic effects after even relatively mild injuries. If confirmed in humans, such subjects may be excluded from, or receive special protection against, activities involving injury risk. Moreover, the treatment and long-term outcome of traumatic brain injuries may benefit from determining the status of cholinergic systems and associated cognitive functions. (PsycINFO Database Record (c) 2019 APA, all rights reserved).


Assuntos
Concussão Encefálica/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Proteínas de Membrana Transportadoras/metabolismo , Acetilcolina/metabolismo , Animais , Atenção/fisiologia , Colinérgicos , Neurônios Colinérgicos/fisiologia , Cognição/fisiologia , Feminino , Masculino , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Transmissão Sináptica/fisiologia
15.
Neurobiol Learn Mem ; 158: 60-72, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30611884

RESUMO

Memory impairments and cognitive decline persist long after recovery from major illness or injury, and correlate with increased risk of later dementia. Here we developed a subchronic peripheral immune challenge model to examine delayed and persistent memory impairments in females and in males. We show that intermittent injections of either lipopolysaccharides or Poly I:C cause memory decline in both sexes that are evident eight weeks after the immune challenge. Importantly, we observed sex-specific patterns of deficits. Females showed impairments in object recognition one week after challenge that persisted for at least eight weeks. In contrast, males had intact memory one week after the immune challenge but exhibited broad impairments in memory tasks including object recognition, and both context and tone fear conditioning several months later. The differential patterns of memory deficits in males and in females were observed without sustained microglial activation or changes in blood-brain barrier permeability. Together, these data suggest that transient neuroimmune activity results in differential vulnerabilities of females and males to memory decline after immune challenge. This model will be an important tool for determining the mechanisms in both sexes that contribute to memory impairments that develop over the weeks and months after recovery from illness. Future studies using this model will provide new insights into the role of chronic inflammation in the pathogenesis of long-lasting memory decline and dementias.


Assuntos
Condicionamento Clássico/fisiologia , Transtornos da Memória/imunologia , Transtornos da Memória/fisiopatologia , Reconhecimento Psicológico/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Condicionamento Clássico/efeitos dos fármacos , Modelos Animais de Doenças , Medo/efeitos dos fármacos , Medo/fisiologia , Feminino , Fatores Imunológicos/imunologia , Fatores Imunológicos/farmacologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Masculino , Transtornos da Memória/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/imunologia , Poli I-C/farmacocinética , Reconhecimento Psicológico/efeitos dos fármacos , Fatores Sexuais
16.
Neuropsychopharmacology ; 44(2): 399-407, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29786067

RESUMO

When memories are retrieved they become labile, and subject to alteration by a process known as reconsolidation. Disruption of memory reconsolidation decreases the performance of learned responses, which is often attributed to erasure of the memory; in the case of Pavlovian learning, to a loss of the association between a conditioned stimulus (CS) and unconditioned stimulus (US). However, an alternative interpretation is that disrupting reconsolidation does not erase memories, but blunts their emotional/motivational impact. It is difficult to parse the predictive vs. emotional/motivational value of CSs in non-human animals, but studies on variation in the form of conditioned responses (CRs) in a Pavlovian conditioned approach task suggest a way to do this. In this task a lever-CS paired with a food reward (US) acquires predictive value in all rats, but is attributed with emotional/motivational value to a greater extent in some rats (sign-trackers) than others (goal-trackers). We report that the post-retrieval administration of propranolol selectively attenuates a sign-tracking CR, and the associated neural activation of brain "motive circuits", while having no effect on conditioned orienting behavior in sign-trackers, or on goal-tracking CRs evoked by either a lever-CS or a tone-CS. We conclude that the disruption of reconsolidation by post-retrieval propranolol degrades the emotional/motivational impact of the CS, required for sign-tracking, but leaves the CS-US association intact. The possibility that post-retrieval interventions can reduce the emotional/motivational aspects of memories, without actually erasing them, has important implications for treating maladaptive memories that contribute to some psychiatric disorders.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Emoções/efeitos dos fármacos , Consolidação da Memória/efeitos dos fármacos , Motivação/efeitos dos fármacos , Propranolol/farmacologia , Animais , Sinais (Psicologia) , Masculino , Nadolol/farmacologia , Ratos , Ratos Sprague-Dawley , Recompensa
17.
Front Syst Neurosci ; 12: 39, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30210310

RESUMO

Neuroimmune signaling is increasingly identified as a critical component of neuronal processes underlying memory, emotion and cognition. The interactions of microglia and astrocytes with neurons and synapses, and the individual cytokines and immune signaling molecules that mediate these interactions are a current focus of much research. Here, we discuss neuroimmune activation as a mechanism triggering different states that modulate cognitive and affective processes to allow for appropriate behavior during and after illness or injury. We propose that these states lie on a continuum from a naïve homeostatic baseline state in the absence of stimulation, to acute neuroimmune activity and chronic activation. Importantly, consequences of illness or injury including cognitive deficits and mood impairments can persist long after resolution of immune signaling. This suggests that neuroimmune activation also results in an enduring shift in the homeostatic baseline state with long lasting consequences for neural function and behavior. Such different states can be identified in a multidimensional way, using patterns of cytokine and glial activation, behavioral and cognitive changes, and epigenetic signatures. Identifying distinct neuroimmune states and their consequences for neural function will provide a framework for predicting vulnerability to disorders of memory, cognition and emotion both during and long after recovery from illness.

18.
Curr Opin Behav Sci ; 23: 92-97, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30083579

RESUMO

Recent work on sex differences in learning and memory has demonstrated that females and males differ in cognitive and behavioral strategies, as well as neural mechanisms required to learn, retrieve and express memory. Although our understanding of the mechanisms of memory is highly sophisticated, this work is based on male animals. As such, the study of female memory is narrowed to a comparison with behavior and mechanisms defined in males, resulting in findings of male-specific mechanisms but little understanding of how females learn and store information. In this paper, we discuss a female-focused framework and experimental approaches to deepen our understanding of the strategies and neural mechanisms engaged by females (and males) in learning, consolidation, and retrieval of memory.

19.
Neuropsychopharmacology ; 42(2): 397-407, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27577601

RESUMO

Anxiety disorders are commonly associated with increased generalization of fear from a stress- or trauma-associated environment to a neutral context or environment. Differences in context-associated memory in males and females may contribute to increased susceptibility to anxiety disorders in women. Here we examined sex differences in context fear generalization and its neural correlates. We observed stronger context fear conditioning and more generalization of fear to a similar context in females than males. In addition, context preexposure increased fear conditioning in males and decreased generalization in females. Accordingly, males showed stronger cFos activity in dorsal hippocampus during memory retrieval and context generalization, whereas females showed preferential recruitment of basal amygdala. Together, these findings are consistent with previous research showing that hippocampal activity correlates with reduced context fear generalization. Differential competition between hippocampus and amygdala-dependent processes may thus contribute to sex differences in retrieval of context fear and greater generalization of fear-associated memory.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Generalização Psicológica/fisiologia , Hipocampo/fisiologia , Rememoração Mental/fisiologia , Caracteres Sexuais , Animais , Condicionamento Clássico , Feminino , Masculino , Consolidação da Memória/fisiologia , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fos/metabolismo
20.
J Neurosci Res ; 95(1-2): 472-486, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27870428

RESUMO

The neuroimmune system is significantly sexually dimorphic, with sex differences evident in the number and activation states of microglia, in the activation of astrocytes, and in cytokine release and function. Neuroimmune cells and signaling are now recognized as critical for many neural functions throughout the life span, including synaptic plasticity and memory function. Here we address the question of how cytokines, astrocytes, and microglia contribute to memory, and specifically how neuroimmune modulation of memory differentially affects males and females. Understanding sex differences in both normal memory processes and dysregulation of memory in psychiatric and neurological disorders is critical for developing treatment and preventive strategies for memory disorders that are effective for both men and women. © 2016 Wiley Periodicals, Inc.


Assuntos
Encéfalo , Citocinas/metabolismo , Sistema Imunitário/fisiologia , Memória/fisiologia , Neuroimunomodulação/fisiologia , Caracteres Sexuais , Animais , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/metabolismo , Humanos , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Transtornos Mentais/complicações , Doenças do Sistema Nervoso/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...