Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Eur J Psychotraumatol ; 15(1): 2364441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38973398

RESUMO

Background: Trauma-focused treatments for post-traumatic stress disorder (PTSD) are effective for many patients. However, relapse may occur when acquired extinction memories fail to generalize beyond treatment contexts. A subgroup of PTSD patients - potentially with substantial exposure to early-life adversity (ELA) - show dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which results in lower cortisol levels. Glucocorticoids, including cortisol, appear to facilitate strength and generalization of emotional memories.Objective: We describe the protocol of an integrated PTSD study. We investigate (A) associations between HPA-axis dysregulation, ELA, epigenetic markers, and PTSD treatment outcome (observational study); and (B) effects of exogenous glucocorticoids on strength and generalization of extinction memories and associated neural mechanisms [pharmacological intervention study with functional magnetic resonance imaging (fMRI)]. The objective is to provide proof of concept that PTSD patients with HPA-axis dysregulation often experienced ELA and may show improved strength and generalization of extinction learning after glucocorticoid administration.Method: The observational study (n = 160 PTSD group, n = 30 control group) assesses ELA, follow-up PTSD symptoms, epigenetic markers, and HPA-axis characteristics (salivary cortisol levels during low-dose dexamethasone suppression test and socially evaluated cold-pressor test). The pharmacological intervention study (n = 80 PTSD group, with and without HPA-axis dysregulation) is a placebo-controlled fMRI study with a crossover design. To investigate strength and generalization of extinction memories, we use a differential fear acquisition, extinction, and extinction recall task with spatial contexts within a virtual environment. Prior to extinction learning, 20 mg hydrocortisone or placebo is administered. During next-day recall, strength of the extinction memory is determined by recovery of skin conductance and pupil dilation differential responding, whereas generalization is assessed by comparing responses between different spatial contexts.Conclusion: The integrated study described in the current protocol paper could inform a personalized treatment approach in which these PTSD patients may receive glucocorticoids as a treatment enhancer in trauma-focused therapies.Trial registration: The research project is registered in the European Union Drug Regulating Authorities Clinical Trials (EudraCT) database, https://eudract.ema.europa.eu/, EudraCT number 2020-000712-30.


This protocol reports a proof-of-concept study for glucocorticoids as an enhancer for PTSD treatment.The study examines whether glucocorticoids enhance the strength and generalization of extinction memory.A further aim is to identify HPA-axis-related PTSD subgroups that may particularly benefit.


Assuntos
Extinção Psicológica , Glucocorticoides , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Transtornos de Estresse Pós-Traumáticos , Humanos , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Glucocorticoides/farmacologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Hidrocortisona , Masculino , Adulto , Feminino , Imageamento por Ressonância Magnética
2.
Nat Commun ; 14(1): 6037, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37758725

RESUMO

Memories undergo a time-dependent neural reorganization, which is assumed to be accompanied by a transformation from detailed to more gist-like memory. However, the nature of this transformation and its underlying neural mechanisms are largely unknown. Here, we report that the time-dependent transformation of memory is semantic in nature, while we find no credible evidence for a perceptual transformation. Model-based MRI analyses reveal time-dependent increases in semantically transformed representations of events in prefrontal and parietal cortices, while specific pattern representations in the anterior hippocampus decline over time. Posterior hippocampal memory reinstatement, in turn, increases over time and is linked to the semantic gist of the original memory, without a statistically significant link to perceptual details. These findings indicate that qualitative changes in memory over time, associated with distinct representational changes in the neocortex and within the hippocampus, reflect a semantic transformation, which may promote the integration of memories into abstract knowledge structures.


Assuntos
Memória , Neocórtex , Humanos , Hipocampo , Neocórtex/diagnóstico por imagem , Lobo Parietal , Semântica
3.
Neurobiol Learn Mem ; 197: 107700, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36410654

RESUMO

Previous findings have indicated that glucocorticoid hormones impair working memory via an interaction with the ß-adrenoceptor-cAMP signaling cascade to rapidly increase cAMP-dependent protein kinase (PKA) activity within the prefrontal cortex (PFC). However, it remains elusive how such activation of PKA can affect downstream cellular mechanisms in regulating PFC cognitive function. PKA is known to activate l-type voltage-gated Ca2+ channels (LTCCs) which regulate a broad range of cellular processes, including neuronal excitability and neurotransmitter release. The present experiments examined whether LTCC activity within the PFC is required in mediating glucocorticoid and PKA effects on spatial working memory. Male Sprague Dawley rats received bilateral administration of the LTCC inhibitor diltiazem together with either the glucocorticoid receptor agonist RU 28362 or PKA activator Sp-cAMPS into the PFC before testing on a delayed alternation task in a T-maze. Both RU 28362 and Sp-cAMPS impaired working memory, whereas the LTCC inhibitor diltiazem fully blocked the working memory impairment induced by either RU 28362 or Sp-cAMPS. Conversely, bilateral administration of the LTCC agonist Bay K8644 into the PFC was sufficient to impair working memory. Thus, these findings provide support for the view that glucocorticoids, via an interaction with the ß-adrenergic signaling cascade and enhanced PKA activity levels, impair working memory by increasing LTCC activity in the PFC.


Assuntos
Glucocorticoides , Memória de Curto Prazo , Ratos , Animais , Masculino , Memória de Curto Prazo/fisiologia , Glucocorticoides/farmacologia , Canais de Cálcio Tipo L/metabolismo , Ratos Sprague-Dawley , Diltiazem/metabolismo , Diltiazem/farmacologia , Transtornos da Memória , Córtex Pré-Frontal/fisiologia
4.
Proc Natl Acad Sci U S A ; 119(41): e2204900119, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-36191198

RESUMO

Emotional information is better remembered than neutral information. Extensive evidence indicates that the amygdala and its interactions with other cerebral regions play an important role in the memory-enhancing effect of emotional arousal. While the cerebellum has been found to be involved in fear conditioning, its role in emotional enhancement of episodic memory is less clear. To address this issue, we used a whole-brain functional MRI approach in 1,418 healthy participants. First, we identified clusters significantly activated during enhanced memory encoding of negative and positive emotional pictures. In addition to the well-known emotional memory-related cerebral regions, we identified a cluster in the cerebellum. We then used dynamic causal modeling and identified several cerebellar connections with increased connection strength corresponding to enhanced emotional memory, including one to a cluster covering the amygdala and hippocampus, and bidirectional connections with a cluster covering the anterior cingulate cortex. The present findings indicate that the cerebellum is an integral part of a network involved in emotional enhancement of episodic memory.


Assuntos
Nível de Alerta , Emoções , Tonsila do Cerebelo , Mapeamento Encefálico , Cerebelo , Humanos , Imageamento por Ressonância Magnética , Rememoração Mental
5.
Epigenomics ; 14(15): 887-895, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-36004496

RESUMO

Epigenomic and neurocognitive studies have provided new perspectives on post-traumatic stress disorder and its intergenerational transmission. This article outlines the lessons learned from community engagement (CE) in such research on Rwandan genocide survivors. A strong trauma-related response was observed within the research project-targeted community (genocide survivors) during explanation of the project. CE also revealed privacy concerns, as community members worried that any leakage of genetic/(epi)genomic data could affect not only themselves but also their close relatives. Adopting a culture of CE in the process of research implementation enables the prioritization of targeted community needs and interests. Furthermore, CE has stimulated the development of mental healthcare interventions, which married couples can apply to protect their offspring and thus truly break the cycle of inherited vulnerability.


Studies of how human genes are affected by the environment (epigenomic studies) have provided new perspectives on post-traumatic stress disorder and its intergenerational transmission. This article describes the lessons learned from community engagement (CE) in this type of research in a Rwandan genocide-exposed population. A strong trauma-related response was observed within the community while explaining the project. CE also revealed the participants' privacy concerns related to leakage of genetic/(epi)genomic data that could also affect their close relatives. Adopting a culture of CE in the process of research implementation enables the prioritization of community needs and interests. CE has furthermore stimulated the development of preventive interventions for married couples to protect their offspring and thus truly break the cycle of inherited vulnerability.


Assuntos
Genocídio , Transtornos de Estresse Pós-Traumáticos , Epigenômica , Genocídio/psicologia , Humanos , Ruanda , Transtornos de Estresse Pós-Traumáticos/genética , Sobreviventes/psicologia
6.
Proc Natl Acad Sci U S A ; 119(22): e2203680119, 2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35622887

RESUMO

Noradrenergic activation of the basolateral amygdala (BLA) by emotional arousal enhances different forms of recognition memory via functional interactions with the insular cortex (IC). Human neuroimaging studies have revealed that the anterior IC (aIC), as part of the salience network, is dynamically regulated during arousing situations. Emotional stimulation first rapidly increases aIC activity but suppresses it in a delayed fashion. Here, we investigated in male Sprague-Dawley rats whether the BLA influence on recognition memory is associated with an increase or suppression of aIC activity during the postlearning consolidation period. We first employed anterograde and retrograde viral tracing and found that the BLA sends dense monosynaptic projections to the aIC. Memory-enhancing norepinephrine administration into the BLA following an object training experience suppressed aIC activity 1 h later, as determined by a reduced expression of the phosphorylated form of the transcription factor cAMP response element-binding (pCREB) protein and neuronal activity marker c-Fos. In contrast, the number of perisomatic γ-aminobutyric acid (GABA)ergic inhibitory synapses per pCREB-positive neuron was significantly increased, suggesting a dynamic up-regulation of GABAergic tone. In support of this possibility, pharmacological inhibition of aIC activity with a GABAergic agonist during consolidation enhanced object recognition memory. Norepinephrine administration into the BLA did not affect neuronal activity within the posterior IC, which receives sparse innervation from the BLA. The evidence that noradrenergic activation of the BLA enhances the consolidation of object recognition memory via a mechanism involving a suppression of aIC activity provides insight into the broader brain network dynamics underlying emotional regulation of memory.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Emoções , Córtex Insular , Inibição Neural , Reconhecimento Psicológico , Percepção Visual , Animais , Nível de Alerta , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/fisiologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Emoções/efeitos dos fármacos , Emoções/fisiologia , Agonistas GABAérgicos/farmacologia , Córtex Insular/efeitos dos fármacos , Córtex Insular/fisiologia , Masculino , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Norepinefrina/administração & dosagem , Norepinefrina/farmacologia , Ratos , Ratos Sprague-Dawley , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Percepção Visual/fisiologia
7.
Neuron ; 110(9): 1450-1467, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35316661

RESUMO

It is well established that stress has a major impact on memory, driven by the concerted action of various stress mediators on the brain. Recent years, however, have seen considerable advances in our understanding of the cellular, neural network, and cognitive mechanisms through which stress alters memory. These novel insights highlight the intricate interplay of multiple stress mediators, including-beyond corticosteroids, catecholamines, and peptides-for instance, endocannabinoids, which results in time-dependent shifts in large-scale neural networks. Such stress-induced network shifts enable highly specific memories of the stressful experience in the long run at the cost of transient impairments in mnemonic flexibility during and shortly after a stressful event. Based on these recent discoveries, we provide a new integrative framework that links the cellular, systems, and cognitive mechanisms underlying acute stress effects on memory processes and points to potential targets for treating aberrant memory in stress-related mental disorders.


Assuntos
Encéfalo , Memória , Catecolaminas , Humanos
8.
Eur J Neurosci ; 55(9-10): 2666-2683, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-33840130

RESUMO

Glucocorticoids enhance memory consolidation of emotionally arousing events via largely unknown molecular mechanisms. This glucocorticoid effect on the consolidation process also requires central noradrenergic neurotransmission. The intracellular pathways of these two stress mediators converge on two transcription factors: the glucocorticoid receptor (GR) and phosphorylated cAMP response element-binding protein (pCREB). We therefore investigated, in male rats, whether glucocorticoid effects on memory are associated with genomic interactions between the GR and pCREB in the hippocampus. In a two-by-two design, object exploration training or no training was combined with post-training administration of a memory-enhancing dose of corticosterone or vehicle. Genomic effects were studied by chromatin immunoprecipitation followed by sequencing (ChIP-seq) of GR and pCREB 45 min after training and transcriptome analysis after 3 hr. Corticosterone administration induced differential GR DNA-binding and regulation of target genes within the hippocampus, largely independent of training. Training alone did not result in long-term memory nor did it affect GR or pCREB DNA-binding and gene expression. No strong evidence was found for an interaction between GR and pCREB. Combination of the GR DNA-binding and transcriptome data identified a set of novel, likely direct, GR target genes that are candidate mediators of corticosterone effects on memory consolidation. Cell-specific expression of the identified target genes using single-cell expression data suggests that the effects of corticosterone reflect in part non-neuronal cells. Together, our data identified new GR targets associated with memory consolidation that reflect effects in both neuronal and non-neuronal cells.


Assuntos
Glucocorticoides , Consolidação da Memória , Animais , Corticosterona/metabolismo , Corticosterona/farmacologia , DNA/metabolismo , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Hipocampo/metabolismo , Masculino , Ratos , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
9.
Eur J Neurosci ; 55(9-10): 2122-2141, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34812558

RESUMO

Abundant evidence shows that early-life stress (ELS) predisposes for the development of stress-related psychopathology when exposed to stressors later in life, but the underlying mechanisms remain unclear. To study predisposing effects of mild ELS on stress sensitivity, we examined in a healthy human population the impact of a history of ELS on acute stress-related changes in corticolimbic circuits involved in emotional processing (i.e., amygdala, hippocampus and ventromedial prefrontal cortex [vmPFC]). Healthy young male participants (n = 120) underwent resting-state functional magnetic resonance imaging (fMRI) in two separate sessions (stress induction vs. control). The Childhood Trauma Questionnaire (CTQ) was administered to index self-reported ELS, and stress induction was verified using salivary cortisol, blood pressure, heart rate and subjective affect. Our findings show that self-reported ELS was negatively associated with baseline cortisol, but not with the acute stress-induced cortisol response. Critically, individuals with more self-reported ELS exhibited an exaggerated reduction of functional connectivity in corticolimbic circuits under acute stress. A mediation analysis showed that the association between ELS and stress-induced changes in amygdala-hippocampal connectivity became stronger when controlling for basal cortisol. Our findings show, in a healthy sample, that the effects of mild ELS on functioning of corticolimbic circuits only become apparent when exposed to an acute stressor and may be buffered by adaptations in hypothalamic-pituitary-adrenal axis function. Overall, our findings might reveal a potential mechanism whereby even mild ELS might confer vulnerability to exposure to stressors later in adulthood.


Assuntos
Experiências Adversas da Infância , Adulto , Humanos , Hidrocortisona , Sistema Hipotálamo-Hipofisário , Imageamento por Ressonância Magnética , Masculino , Sistema Hipófise-Suprarrenal , Estresse Psicológico
10.
Epigenomics ; 14(1): 11-25, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34875875

RESUMO

Aim & methods: We conducted a pilot epigenome-wide association study of women from Tutsi ethnicity exposed to the genocide while pregnant and their resulting offspring, and a comparison group of women who were pregnant at the time of the genocide but living outside of Rwanda.Results: Fifty-nine leukocyte-derived DNA samples survived quality control: 33 mothers (20 exposed, 13 unexposed) and 26 offspring (16 exposed, 10 unexposed). Twenty-four significant differentially methylated regions (DMRs) were identified in mothers and 16 in children. Conclusions:In utero genocide exposure was associated with CpGs in three of the 24 DMRs: BCOR, PRDM8 and VWDE, with higher DNA methylation in exposed versus unexposed offspring. Of note, BCOR and VWDE show significant correlation between brain and blood DNA methylation within individuals, suggesting these peripherally derived signals of genocide exposure may have relevance to the brain.


Lay abstract The 1994 Rwandan genocide against ethnic Tutsi has been associated with adverse mental health outcomes in survivors decades later, but the molecular mechanisms that contribute to this association remain poorly characterized. Epigenetic mechanisms such as DNA methylation regulate gene function and change in response to life experiences. We identified differentially methylated regions (DMRs) in genocide-exposed versus unexposed mothers and children. In utero genocide exposure was linked with methylation differences in three maternal DMRs, with higher methylation in exposed offspring. Two of three DMRs show correlation between brain and blood methylation within individuals, suggesting that peripherally derived signals of genocide exposure may be relevant to the brain.


Assuntos
Genocídio , Transtornos de Estresse Pós-Traumáticos , Criança , Metilação de DNA , Epigenoma , Feminino , Humanos , Leucócitos , Gravidez , Ruanda , Sobreviventes
11.
Nat Commun ; 12(1): 6054, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663784

RESUMO

It is commonly assumed that episodic memories undergo a time-dependent systems consolidation process, during which hippocampus-dependent memories eventually become reliant on neocortical areas. Here we show that systems consolidation dynamics can be experimentally manipulated and even reversed. We combined a single pharmacological elevation of post-encoding noradrenergic activity through the α2-adrenoceptor antagonist yohimbine with fMRI scanning both during encoding and recognition testing either 1 or 28 days later. We show that yohimbine administration, in contrast to placebo, leads to a time-dependent increase in hippocampal activity and multivariate encoding-retrieval pattern similarity, an indicator of episodic reinstatement, between 1 and 28 days. This is accompanied by a time-dependent decrease in neocortical activity. Behaviorally, these neural changes are linked to a reduced memory decline over time after yohimbine intake. These findings indicate that noradrenergic activity shortly after encoding may alter and even reverse systems consolidation in humans, thus maintaining vividness of memories over time.


Assuntos
Nível de Alerta , Hipocampo/efeitos dos fármacos , Norepinefrina/farmacologia , Ioimbina/farmacologia , Adulto , Método Duplo-Cego , Feminino , Hipocampo/fisiologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Consolidação da Memória/efeitos dos fármacos , Memória Episódica , Memória de Longo Prazo/efeitos dos fármacos , Memória de Longo Prazo/fisiologia , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Adulto Jovem
12.
Neurobiol Learn Mem ; 183: 107481, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34166790

RESUMO

Glucocorticoid hormones are crucially involved in modulating mnemonic processing of stressful or emotionally arousing experiences. They are known to enhance the consolidation of new memories, including those that extinguish older memories. In this study, we investigated whether glucocorticoids facilitate the extinction of a striatum-dependent, and behaviorally more rigid, stimulus-response memory. For this, male rats were initially trained for six days on a stimulus-response task in a T-maze to obtain a reward after making an egocentric right-turn body response, regardless of the starting position in this maze. This training phase was followed by three extinction sessions in which right-turn body responses were not reinforced. Corticosterone administration into the dorsolateral region of the striatum after the first extinction session dose-dependently enhanced the consolidation of extinction memory: Rats administered the higher dose of corticosterone (30 ng), but not lower doses (5 or 10 ng), exhibited significantly fewer right-turn body responses and had longer latencies compared to vehicle-treated animals on the second and third extinction sessions. Co-administration of the glucocorticoid receptor antagonist RU 486 (10 ng) prevented the corticosterone effect, indicating that glucocorticoids enhance the extinction of stimulus-response memory via activation of the glucocorticoid receptor. Corticosterone administration into the dorsomedial striatum did not affect extinction memory. These findings indicate that stress-response mechanisms involving corticosterone actions in the dorsolateral striatum facilitate the extinction of stimulus-response memory that might allow for the development of an opportune behavioral strategy.


Assuntos
Corticosterona/farmacologia , Extinção Psicológica/efeitos dos fármacos , Glucocorticoides/farmacologia , Memória/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Antagonistas de Hormônios/farmacologia , Masculino , Aprendizagem em Labirinto , Consolidação da Memória/efeitos dos fármacos , Mifepristona/farmacologia , Neostriado/metabolismo , Neostriado/patologia , Ratos , Receptores de Glucocorticoides/antagonistas & inibidores
13.
Stress ; 24(2): 181-188, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32233890

RESUMO

Extensive evidence indicates that noradrenergic activation is essentially involved in mediating the enhancing effects of emotional arousal on memory consolidation. Our current understanding of the neurobiological mechanisms underlying the memory-modulatory effects of the noradrenergic system is primarily based on pharmacological studies in rats, employing targeted administration of noradrenergic drugs into specific brain regions. However, the further delineation of the specific neural circuitry involved would benefit from experimental tools that are currently more readily available in mice. Previous studies have not, as yet, investigated the effect of noradrenergic enhancement of memory in mice, which show different cognitive abilities and higher endogenous arousal levels induced by a training experience compared to rats. In the present study, we investigated the effect of posttraining noradrenergic activation in male C57BL/6J mice on the consolidation of object recognition and object location memory. We found that the noradrenergic stimulant yohimbine (0.3 or 1.0 mg/kg) administered systemically immediately after an object training experience dose-dependently enhanced 24-h memory of both the identity and location of the object. Thus, these findings indicate that noradrenergic activation also enhances memory consolidation processes in mice, paving the way for a systematic investigation of the neural circuitry underlying these emotional arousal effects on memory.LAY SUMMARY: The current study successfully validated the effect of noradrenergic activation on both object recognition and object location memory in mice. This study thereby provides a fundamental proof-of-principle for the investigation of the neural circuitry underlying noradrenergic and arousal effects on long-term memory in mice.


Assuntos
Memória , Estresse Psicológico , Animais , Masculino , Memória de Longo Prazo , Camundongos , Camundongos Endogâmicos C57BL , Norepinefrina , Ratos
14.
Mol Cell Neurosci ; 108: 103537, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32805389

RESUMO

Stressful and emotionally arousing experiences activate hormonal and brain systems that create strong memories. Extensive evidence indicates that this strengthening effect involves the synergistic action of both norepinephrine and glucocorticoid hormones. This tight regulation of emotional memories is normally highly adaptive and pivotal for survival; yet, aberrant memory processing of stressful events is a major risk factor for the development of stress-related psychopathology. It remains unclear, however, to what extent these two stress hormone systems also affect the quality of such strengthened memories. In this Review, we discuss recent advances in the understanding of norepinephrine and glucocorticoid effects on the accuracy and generalization of contextual or episodic-like aspects of memory in rodents. We will argue that norepinephrine and glucocorticoids exert opposite effects on accuracy and generalization of memory through distinct effects on systems consolidation processes underlying the time-dependent reorganization of memory. Norepinephrine improves memory accuracy by boosting basolateral amygdala-hippocampal connectivity, hereby creating long-lasting hippocampus-dependent episodic-like memories. In contrast, glucocorticoids contribute to memory generalization by promoting integration of new memories into neocortical networks, decreasing hippocampal dependence. We discuss possible implications of these conceptual insights for understanding inter-individual differences in stress resilience and risk for psychopathology.


Assuntos
Encéfalo/metabolismo , Glucocorticoides/metabolismo , Memória , Norepinefrina/metabolismo , Estresse Psicológico/metabolismo , Animais , Encéfalo/fisiologia , Generalização Psicológica , Humanos , Estresse Psicológico/fisiopatologia
15.
Psychoneuroendocrinology ; 114: 104588, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32085987

RESUMO

Stressful and emotionally arousing experiences activate hormonal systems that create strong memories. It remains unclear, however, how this strengthening affects the quality of such memories. In the present study, we examined whether the noradrenergic and glucocorticoid hormonal systems affect accuracy of episodic-like memory. We trained male Sprague-Dawley rats on an episodic-like association task, termed inhibitory avoidance discrimination task, in which they explored two different contexts, but shock was given only in the latter context. Forty-eight hours later, retention latencies were tested in the two training contexts as well as in a novel context. The noradrenergic stimulant yohimbine, administered systemically immediately after the training session, enhanced both accuracy and strength of the memory, as shown by long latencies specific to the shock context. By contrast, the glucocorticoid corticosterone induced a generalized strengthening of memory and enhanced latencies in both the shock and non-shock training contexts. Retention latencies in the novel context were not significantly affected. These findings indicate that the noradrenergic and glucocorticoid systems, while both strengthening memory of the shock experience per se, produce opposite effects on accuracy of the shock-context association.


Assuntos
Condicionamento Clássico/fisiologia , Glucocorticoides/fisiologia , Memória Episódica , Norepinefrina/fisiologia , Desempenho Psicomotor/fisiologia , Retenção Psicológica/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Condicionamento Clássico/efeitos dos fármacos , Corticosterona/farmacologia , Aprendizagem por Discriminação/efeitos dos fármacos , Aprendizagem por Discriminação/fisiologia , Glucocorticoides/farmacologia , Inibição Psicológica , Masculino , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Retenção Psicológica/efeitos dos fármacos , Ioimbina/farmacologia
16.
Front Mol Neurosci ; 12: 292, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31849606

RESUMO

Human studies have consistently shown that drugs of abuse affect memory function. The psychostimulants amphetamine and the "bath salt" 3,4-methylenedioxypyrovalerone (MDPV) increase brain monoamine levels through a similar, yet not identical, mechanism of action. Findings indicate that amphetamine enhances the consolidation of memory for emotional experiences, but still MDPV effects on memory function are underinvestigated. Here, we tested the effects induced by these two drugs on generalization of fear memory and their relative neurobiological underpinnings. To this aim, we used a modified version of the classical inhibitory avoidance task, termed inhibitory avoidance discrimination task. According to such procedure, adult male Sprague-Dawley rats were first exposed to one inhibitory avoidance apparatus and, with a 1-min delay, to a second apparatus where they received an inescapable footshock. Forty-eight hours later, retention latencies were tested, in a randomized order, in the two training apparatuses as well as in a novel contextually modified apparatus to assess both strength and generalization of memory. Our results indicated that both amphetamine and MDPV induced generalization of fear memory, whereas only amphetamine enhanced memory strength. Co-administration of the ß-adrenoceptor antagonist propranolol prevented the effects of both amphetamine and MDPV on the strength and generalization of memory. The dopaminergic receptor blocker cis-flupenthixol selectively reversed the amphetamine effect on memory generalization. These findings indicate that amphetamine and MDPV induce generalization of fear memory through different modulations of noradrenergic and dopaminergic neurotransmission.

17.
J Neuroendocrinol ; 31(8): e12735, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31121060

RESUMO

Brain mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs) respond to the same glucocorticoid hormones but can have differential effects on cellular function. Several lines of evidence suggest that MR-specific target genes must exist and might underlie the distinct effects of the receptors. The present study aimed to identify MR-specific target genes in the hippocampus, a brain region where MR and GR are co-localised and play a role in the stress response. Using genome-wide binding of both receptor types, we previously identified MR-specific, MR-GR overlapping and GR-specific putative target genes. We now report altered gene expression levels of such genes in the hippocampus of forebrain MR knockout (fbMRKO) mice, killed at the time of their endogenous corticosterone peak. Of those genes associated with MR-specific binding, the most robust effect was a 50% reduction in Jun dimerization protein 2 (Jdp2) mRNA levels in fbMRKO mice. Down-regulation was also observed for the MR-specific Nitric oxide synthase 1 adaptor protein (Nos1ap) and Suv3 like RNA helicase (Supv3 l1). Interestingly, the classical glucocorticoid target gene FK506 binding protein 5 (Fkbp5), which is associated with MR and GR chromatin binding, was expressed at substantially lower levels in fbMRKO mice. Subsequently, hippocampal Jdp2 was confirmed to be up-regulated in a restraint stress model, posing Jdp2 as a bona fide MR target that is also responsive in an acute stress condition. Thus, we show that MR-selective DNA binding can reveal functional regulation of genes and further identify distinct MR-specific effector pathways.


Assuntos
Regulação da Expressão Gênica , Hipocampo/metabolismo , Receptores de Mineralocorticoides/fisiologia , Animais , Sítios de Ligação/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/genética
18.
Int J Mol Sci ; 20(7)2019 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-30934833

RESUMO

Mineralocorticoid receptor (MR)-mediated signaling in the brain has been suggested as a protective factor in the development of psychopathology, in particular mood disorders. We recently identified genomic loci at which either MR or the closely related glucocorticoid receptor (GR) binds selectively, and found members of the NeuroD transcription factor family to be specifically associated with MR-bound DNA in the rat hippocampus. We show here using forebrain-specific MR knockout mice that GR binding to MR/GR joint target loci is not affected in any major way in the absence of MR. Neurod2 binding was also independent of MR binding. Moreover, functional comparison with MyoD family members indicates that it is the chromatin remodeling aspect of NeuroD, rather than its direct stimulation of transcription, that is responsible for potentiation of MR-mediated transcription. These findings suggest that NeuroD acts in a permissive way to enhance MR-mediated transcription, and they argue against competition for DNA binding as a mechanism of MR- over GR-specific binding.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transdução de Sinais , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Corticosterona/metabolismo , DNA/metabolismo , Células HEK293 , Hipocampo/metabolismo , Humanos , Masculino , Camundongos Knockout , Modelos Biológicos , Proteína MyoD/metabolismo , Proteínas do Tecido Nervoso/química , Ligação Proteica , Domínios Proteicos , Receptores de Glucocorticoides/metabolismo , Relação Estrutura-Atividade
19.
Proc Natl Acad Sci U S A ; 116(14): 7077-7082, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30877244

RESUMO

Extensive evidence indicates that the basolateral amygdala (BLA) interacts with other brain regions in mediating stress hormone and emotional arousal effects on memory consolidation. Brain activation studies have shown that arousing conditions lead to the activation of large-scale neural networks and several functional connections between brain regions beyond the BLA. Whether such distal interactions on memory consolidation also depend on BLA activity is not as yet known. We investigated, in male Sprague-Dawley rats, whether BLA activity enables prelimbic cortex (PrL) interactions with the anterior insular cortex (aIC) and dorsal hippocampus (dHPC) in regulating glucocorticoid effects on different components of object recognition memory. The glucocorticoid receptor (GR) agonist RU 28362 administered into the PrL, but not infralimbic cortex, immediately after object recognition training enhanced 24-hour memory of both the identity and location of the object via functional interactions with the aIC and dHPC, respectively. Importantly, posttraining inactivation of the BLA by the noradrenergic antagonist propranolol abolished the effect of GR agonist administration into the PrL on memory enhancement of both the identity and location of the object. BLA inactivation by propranolol also blocked the effect of GR agonist administration into the PrL on inducing changes in neuronal activity within the aIC and dHPC during the postlearning consolidation period as well as on structural changes in spine morphology assessed 24 hours later. These findings provide evidence that BLA noradrenergic activity enables functional interactions between the PrL and the aIC and dHPC in regulating stress hormone and emotional arousal effects on memory.


Assuntos
Androstanóis/farmacologia , Complexo Nuclear Basolateral da Amígdala/metabolismo , Córtex Cerebral/metabolismo , Glucocorticoides/metabolismo , Memória/efeitos dos fármacos , Rede Nervosa/metabolismo , Receptores de Glucocorticoides/agonistas , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA