Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(3): 579-586, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28397837

RESUMO

The hippocampus and prefrontal cortex (PFC) are connected in a reciprocal manner: whereas the hippocampus projects directly to the PFC, a polysynaptic pathway that passes through the nucleus reuniens (RE) of the thalamus relays inputs from the PFC to the hippocampus. The present study demonstrates that lesioning and/or inactivation of the RE reduces coherence in the PFC-hippocampal pathway, provokes an antidepressant-like behavioral response in the forced swim test and prevents, but does not ameliorate, anhedonia in the chronic mild stress (CMS) model of depression. Additionally, RE lesioning before CMS abrogates the well-known neuromorphological and endocrine correlates of CMS. In summary, this work highlights the importance of the reciprocal connectivity between the hippocampus and PFC in the establishment of stress-induced brain pathology and suggests a role for the RE in promoting resilience to depressive illness.


Assuntos
Depressão/metabolismo , Núcleos da Linha Média do Tálamo/fisiologia , Estresse Psicológico/metabolismo , Animais , Antidepressivos/metabolismo , Transtorno Depressivo/metabolismo , Hipocampo/fisiologia , Masculino , Núcleos da Linha Média do Tálamo/metabolismo , Vias Neurais/fisiologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Ratos
2.
Mol Psychiatry ; 22(8): 1110-1118, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28555078

RESUMO

Stress, a well-known sculptor of brain plasticity, is shown to suppress hippocampal neurogenesis in the adult brain; yet, the underlying cellular mechanisms are poorly investigated. Previous studies have shown that chronic stress triggers hyperphosphorylation and accumulation of the cytoskeletal protein Tau, a process that may impair the cytoskeleton-regulating role(s) of this protein with impact on neuronal function. Here, we analyzed the role of Tau on stress-driven suppression of neurogenesis in the adult dentate gyrus (DG) using animals lacking Tau (Tau-knockout; Tau-KO) and wild-type (WT) littermates. Unlike WTs, Tau-KO animals exposed to chronic stress did not exhibit reduction in DG proliferating cells, neuroblasts and newborn neurons; however, newborn astrocytes were similarly decreased in both Tau-KO and WT mice. In addition, chronic stress reduced phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR)/glycogen synthase kinase-3ß (GSK3ß)/ß-catenin signaling, known to regulate cell survival and proliferation, in the DG of WT, but not Tau-KO, animals. These data establish Tau as a critical regulator of the cellular cascades underlying stress deficits on hippocampal neurogenesis in the adult brain.


Assuntos
Neurogênese/fisiologia , Proteínas tau/metabolismo , Animais , Astrócitos/metabolismo , Proliferação de Células , Sobrevivência Celular , Giro Denteado/metabolismo , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Transdução de Sinais , Estresse Fisiológico , beta Catenina/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-39162717

RESUMO

RATIONALE: Depression is a serious psychiatric disease, which is diagnosed twice as frequently in women than men. We have recently shown that lesioning or inactivation of the nucleus reuniens (RE), which interconnects the prefrontal cortex (PFC) and hippocampus, promoted resilience to stress in males, exerts an antidepressant effect in the Forced Swim Test (FST) and prevents the development of behavioral and neurobiological alterations induced by the chronic mild stress model of depression. OBJECTIVES: In this study, we expand our findings on the FST in female rats and we investigate whether RE lesion presents sex differences following treatment with two distinct antidepressants, a selective serotonin reuptake inhibitor, i.e. sertraline and a tricyclic antidepressant, i.e. clomipramine. METHODS: Male and female rats received either a surgical lesion of the RE or sham operation, then treated with vehicle, sertraline (10mg/kg) or clomipramine (10mg/kg) and were subjected to the FST. Activation of key brain areas of interest (PFC, Hippocampus and RE) were measured by c-Fos immunoreactivity. RESULTS: RE lesion induced an antidepressant-like phenotype in both female and male rats, confirming its crucial role in the stress response. Similarly to RE lesion, sertraline treatment resulted in increased swimming and decreased immobility duration, as well as enhanced head shake frequency, in both sexes. Notably, climbing behavior was increased only following clomipramine treatment. RE area was less active in females compared to male rats and in clomipramine-treated males compared to their corresponding vehicle-group. Activation of the PFC and the CA1 hippocampal area was reduced in clomipramine-treated females, in comparison to vehicle-treated female rats. This effect was not evident in males, which exhibited less activation in the PFC and the hippocampus than females. CONCLUSION: Re lesion proves equally effective in female and male rats, but sex is highlighted as a pivotal factor in behavioral and treatment response in FST, as well as in related circuit connectivity and activation.

4.
Neuroscience ; 454: 140-150, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32512138

RESUMO

Steroid hormones secreted by the gonads (sex steroids) and adrenal glands (glucocorticoids, GC) are known to influence brain structure and function. While levels of sex steroids wane in late adulthood, corticosteroid levels tend to rise in many individuals due to age-related impairments in their feedback on central mechanisms regulating adrenal function. These fluctuations in sex and adrenal steroid secretion may be relevant to age-related neurodegenerative disorders such as Alzheimer's disease (AD) in which hyperphosphorylation of Tau protein is a key pathological event. We here report that both, long-term GC deprivation (by adrenalectomy) and exogenous GC administration with natural or synthetic glucocorticoid receptor ligands (corticosterone and dexamethasone, respectively) induce Tau hyperphosphorylation in the hippocampus and frontocortical regions at epitopes associated with disruption of cytoskeletal and synaptic function. Interestingly, we observed that the changes in Tau induced by manipulation of the GC milieu of male rats were exacerbated by testosterone depletion (by orchiectomy). While this finding supports previous suggestions of a neuroprotective role of male sex hormones, this is the first study to address interactions between adrenal and sex steroids on Tau hyperphosphorylation and accumulation that are known to endanger neuronal function and plasticity. These results are particularly important for understanding the mechanisms that can precipitate AD because, besides being modulated by age, GC are elevated by stress, a phenomenon now established as a trigger of deficits in neural plasticity and survival, cognitive behaviour and AD-like Tau pathology.


Assuntos
Doença de Alzheimer , Proteínas tau , Animais , Encéfalo/metabolismo , Glucocorticoides , Hormônios Esteroides Gonadais , Hipocampo/metabolismo , Masculino , Fosforilação , Ratos , Proteínas tau/metabolismo
5.
Mol Psychiatry ; 14(1): 95-105, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17912249

RESUMO

Observations of elevated basal cortisol levels in Alzheimer's disease (AD) patients prompted the hypothesis that stress and glucocorticoids (GC) may contribute to the development and/or maintenance of AD. Consistent with that hypothesis, we show that stress and GC provoke misprocessing of amyloid precursor peptide in the rat hippocampus and prefrontal cortex, resulting in increased levels of the peptide C-terminal fragment 99 (C99), whose further proteolytic cleavage results in the generation of amyloid-beta (Abeta). We also show that exogenous Abeta can reproduce the effects of stress and GC on C99 production and that a history of stress strikingly potentiates the C99-inducing effects of Abeta and GC. Previous work has indicated a role for Abeta in disruption of synaptic function and cognitive behaviors, and AD patients reportedly show signs of heightened anxiety. Here, behavioral analysis revealed that like stress and GC, Abeta administration causes spatial memory deficits that are exacerbated by stress and GC; additionally, Abeta, stress and GC induced a state of hyperanxiety. Given that the intrinsic properties of C99 and Abeta include neuroendangerment and behavioral impairment, our findings suggest a causal role for stress and GC in the etiopathogenesis of AD, and demonstrate that stressful life events and GC therapy can have a cumulative impact on the course of AD development and progression.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Comportamento Animal/fisiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Modelos Animais de Doenças , Emoções/fisiologia , Glucocorticoides/sangue , Hipocampo/metabolismo , Masculino , Memória/fisiologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Wistar , Percepção Espacial/fisiologia , Estresse Psicológico/patologia
6.
Eur Neuropsychopharmacol ; 29(1): 1-15, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30497839

RESUMO

Environmental enrichment (EE), comprising positive physical (exercise) and cognitive stimuli, influences neuronal structure and usually improves brain function. The promise of EE as a preventative strategy against neuropsychiatric disease is especially high during early postnatal development when the brain is still amenable to reorganization. Despite the fact that male and female brains differ in terms of connectivity and function that may reflect early life experiences, knowledge of the neural substrates and mechanisms by which such changes arise remains limited. This study compared the impact of EE combined with physical activity on neuroplasticity and its functional consequences in adult male and female rats; EE was provided during the first 3 months of life and our analysis focused on the hippocampus, an area implicated in cognitive behavior as well as the neuroendocrine response to stress. Both male and female rats reared in EE displayed better object recognition memory than their control counterparts. Interestingly, sex differences were revealed in the effects of EE on time spent exploring the objects during this test. Independently of sex, EE increased hippocampal turnover rates of dopamine and serotonin and reduced expression of 5-HT1A receptors; in addition, EE upregulated expression of synaptophysin, a presynaptic protein, in the hippocampus. As compared to their respective controls, EE-exposed males exhibited parallel increases in phosphorylated Tau and the GluN2B receptor, whereas females responded to EE with reduced hippocampal levels of glutamate and GluN2B. Together, these observations provide further evidence on the differential effects of EE on markers of hippocampal neuroplasticity in males and females.


Assuntos
Cognição/fisiologia , Meio Ambiente , Comportamento Exploratório/fisiologia , Plasticidade Neuronal/fisiologia , Condicionamento Físico Animal/fisiologia , Caracteres Sexuais , Animais , Dopamina/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Fosforilação , Ratos , Receptor 5-HT1A de Serotonina/biossíntese , Receptores de N-Metil-D-Aspartato/metabolismo , Reconhecimento Psicológico/fisiologia , Serotonina/metabolismo , Sinaptofisina/biossíntese , Proteínas tau/metabolismo
7.
Neurosci Biobehav Rev ; 32(6): 1161-73, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18573532

RESUMO

Increasingly, stress is recognized as a trigger of depressive episodes and recent evidence suggests a causal role of stress in the onset and progression of Alzheimer's disease (AD) pathology. Besides aging, sex is an important determinant of prevalence rates for both AD and mood disorders. In light of a recent meta-analysis indicating that depressed subjects have a higher likelihood of developing AD, a key message in this article will be that both depression and AD are stress-related disorders and may represent a continuum that should receive more attention in future neurobiological studies. Accordingly, this review considers some of the cellular mechanisms that may be involved in regulating this transition threshold. In addition, it highlights the importance of addressing the question of how aging and sex interplay with stress to influence mood and cognition, with a bias towards consideration of neuroplastic events in particular brain regions, as the basis of AD and depressive disorders.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Transtorno Depressivo/complicações , Glucocorticoides/metabolismo , Estresse Fisiológico/complicações , Doença de Alzheimer/etiologia , Animais , Transtorno Depressivo/patologia , Humanos , Estresse Fisiológico/patologia
8.
Neuroscience ; 152(3): 656-69, 2008 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-18291594

RESUMO

Mood disorders are the most common psychiatric disorders. Although the mechanisms implicated in the genesis of mood disorders are still unclear, stress is known to predispose to depression, and recently, studies have related hippocampal neurogenesis and apoptosis to depression. In the present study we first examined the balance between cell birth-death in the hippocampus and subventricular zone (SVZ) of pre-pubertal and adult rats subjected to chronic-mild-stress (CMS). CMS led to increased corticosterone secretion and induced depressive-like symptoms (assessed in the forced-swimming test); these endocrine and behavioral effects were paralleled by decreased hippocampal, but not SVZ, cell proliferation/differentiation and by increased apoptotic rate. In order to determine if lithium, a known mood stabilizer with antidepressant properties, could prevent the stress-induced events, we analyzed the same parameters in a group of rats treated with lithium during the stress exposure period (CMS+Li) and observed that the hormonal, behavioral and cell turnover effects of CMS were abrogated in these animals. Subsequently, to search for possible pathways through which CMS and lithium influence behavior, cell fate and synaptic plasticity, we analyzed the expression of glycogen-synthase-kinase-3beta (GSK-3beta), as well as some of its downstream targets (B-cell-CLL/lymphoma2-associated athanonege (BAG-1) and synapsin-I). CMS increased GSK-3beta and decreased synapsin-I and BAG-1 expression in the hippocampus. Interestingly, co-administration of lithium precluded the CMS-induced effects in GSK-3beta, synapsin-I and BAG-1 expression. Our observation that specific inhibition of this kinase with AR-A014418 blocked the effects of CMS in depressive-like behavior and in BAG-1 and synapsin-I expression confirmed the involvement of the GSK-3beta pathway in stress-induced effects. In summary, these results reveal that lithium, by regulating the activity of GSK-3beta, prevents the deleterious effects of stress on behavior and cellular functions.


Assuntos
Transtorno Depressivo/tratamento farmacológico , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/efeitos dos fármacos , Cloreto de Lítio/farmacologia , Estresse Psicológico/tratamento farmacológico , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Animais , Antimaníacos/farmacologia , Antimaníacos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Corticosterona/sangue , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Transtorno Depressivo/enzimologia , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Glicogênio Sintase Quinase 3 beta , Hipocampo/citologia , Hipocampo/enzimologia , Cloreto de Lítio/uso terapêutico , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Ratos , Ratos Wistar , Células-Tronco/efeitos dos fármacos , Células-Tronco/enzimologia , Estresse Psicológico/enzimologia , Estresse Psicológico/fisiopatologia , Sinapsinas/efeitos dos fármacos , Sinapsinas/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
Neuroscience ; 454: 1-2, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33478751
10.
J Psychiatr Res ; 39(5): 451-60, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15992553

RESUMO

Imbalances in the corticosteroid milieu result in reductions in hippocampal volume in humans and experimental rodents. The functional correlates of these changes include deficits in cognitive performance and regulation of the hypothalamic-pituitary-adrenal axis. Since other limbic structures which are intricately connected with the hippocampal formation, also play an important role in behavioural and neuroendocrine functions, we here used magnetic resonance imaging (MRI) to analyse how two of these areas, the anterior cingulate and retrosplenial cortex, respond to chronic alterations of adrenocortical status: hypocortisolism (induced by adrenalectomy, ADX), normocortisolism (ADX with low-dose corticosterone replacement), and hypercortisolism (ADX with high-dose dexamethasone supplementation). Hypercortisolism was associated with a significant reduction in the volume (absolute and normalized) of the left anterior cingulate gyrus as measured by MRI and confirmed using classical histological methods; a similar trend was observed in the right anterior cingulate region. In contrast, hypercortisolism did not influence the volume of the adjacent retrosplenial cortex. The volumes of the anterior cingulate gyrus and retrosplenial cortex were unaffected by the absence of adrenocortical hormones. These findings are the first to suggest that corticosteroid influences on the structure of the limbic system extend beyond the hippocampal formation, i.e., to fronto-limbic areas also.


Assuntos
Corticosteroides/fisiologia , Anti-Inflamatórios/farmacologia , Córtex Cerebral/efeitos dos fármacos , Corticosterona/farmacologia , Giro do Cíngulo/efeitos dos fármacos , Adrenalectomia , Animais , Córtex Cerebral/anatomia & histologia , Córtex Cerebral/patologia , Síndrome de Cushing/psicologia , Síndrome de Cushing/veterinária , Giro do Cíngulo/anatomia & histologia , Giro do Cíngulo/patologia , Imageamento por Ressonância Magnética , Masculino , Ratos , Ratos Wistar
11.
Neuroscience ; 194: 62-71, 2011 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-21839808

RESUMO

Disorders such as depression and anxiety exhibit strong sex differences in their prevalence and incidence, with women also differing from men in their response to antidepressants. Furthermore, receptors for corticotrophin releasing hormone (CRHR1) and arginine vasopressin receptor subtype 1b (AVPR1b) are known to contribute to the regulation of mood and anxiety. In the present study, we compared the anxiety profile and CRHR1 and AVPR1b expression levels in control Sprague-Dawley (SD) rats and rats of the SD-derived Flinders Sensitive Line (FSL), a genetic model of depression. Additionally, given the apparent sex differences in the therapeutic efficacy of antidepressants and because antidepressants are commonly used to treat comorbid anxiety and depressive symptoms, we assessed whether the anxiolytic effects of an antidepressant occur in a sex-dependent manner. Male and female FSL rats were treated with citalopram 10 mg/kg once daily for 14 days and were then tested in the open field and the elevated plus maze paradigms. Upon completion of the behavioural analysis, AVPR1b and CRHR1 expression levels were monitored in the hypothalamus and the prefrontal cortex (PFC) using Western blotting. According to our results, male FSL rats were more anxious than control SD rats, a difference abolished by citalopram treatment. Baseline anxiety levels were similar in female FSL and SD rats, and citalopram further reduced anxiety in female FSL rats. Importantly, whereas citalopram altered AVPR1b expression in the hypothalamus of male FSL rats, its actions on this parameter were restricted to the PFC in female FSL rats. In both sexes of FSL rats, citalopram did not alter CRHR1 expression in either the hypothalamus or PFC. Our results demonstrate that antidepressant treatment reduces anxiety levels in FSL rats of both sexes: the magnitude of treatment effect was related to the starting baseline level of anxiety and the antidepressant elicited sexually differentiated neurobiological responses in specific brain regions.


Assuntos
Citalopram/farmacologia , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/genética , Caracteres Sexuais , Animais , Ansiolíticos/farmacologia , Antidepressivos de Segunda Geração/farmacologia , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa