Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Neurosci ; 38(44): 9423-9432, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30381434

RESUMO

Risk for stress-sensitive psychopathologies differs in men and women, yet little is known about sex-dependent effects of stress on cellular structure and function in corticolimbic regions implicated in these disorders. Determining how stress influences these regions in males and females will deepen our understanding of the mechanisms underlying sex-biased psychopathology. Here, we discuss sex differences in CRF regulation of arousal and cognition, glucocorticoid modulation of amygdalar physiology and alcohol consumption, the age-dependent impact of social stress on prefrontal pyramidal cell excitability, stress effects on the prefrontal parvalbumin system in relation to emotional behaviors, contributions of stress and gonadal hormones to stress effects on prefrontal glia, and alterations in corticolimbic structure and function after cessation of chronic stress. These studies demonstrate that, while sex differences in stress effects may be nuanced, nonuniform, and nonlinear, investigations of these differences are nonetheless critical for developing effective, sex-specific treatments for psychological disorders.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Emoções/fisiologia , Motivação/fisiologia , Resiliência Psicológica , Caracteres Sexuais , Estresse Psicológico/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Humanos , Masculino , Transtornos Mentais/metabolismo , Transtornos Mentais/patologia , Transtornos Mentais/psicologia , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fatores de Risco , Estresse Psicológico/patologia , Estresse Psicológico/psicologia
2.
Neurobiol Dis ; 99: 36-46, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27993645

RESUMO

The adolescent brain is marked by functional and structural modifications, particularly within the inhibitory system of the prefrontal cortex (PFC). These changes are necessary for the acquisition of adult cognitive functions and emotion regulation, and impairments in these processes are associated with neuropathologies such as schizophrenia and affective disorders. The molecular mechanisms regulating this adolescent refinement of prefrontal inhibitory circuits remain largely unknown. Here we demonstrate that the transcription factor Npas4 plays a major role in this process. Using a series of behavioral, molecular, pharmacological and genetic approaches in mice, we demonstrate that deficiency in Npas4 affects adolescent expression of multiple markers of GABAergic transmission in the PFC, including parvalbumin and GAD67, in a sex-specific manner. This abnormal pattern of expression of GABAergic markers is associated with sex-specific cognitive and emotional impairments that occur only when Npas4 deficiency begins at adolescence but not post-adolescence. Finally, we show that chronic treatment with the GABA enhancing drug sodium valproate during adolescence is sufficient to induce long-lasting recovery of the molecular and behavioral abnormalities observed in Npas4 deficient mice. Altogether, we provide evidence for the involvement of the transcription factor Npas4 to the structural changes that affect prefrontal inhibitory circuits during adolescence. Further investigations of Npas4 role in the adolescent brain might provide new insights on the molecular mechanisms underlying neuropsychiatric disorders that emerge during adolescence.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cognição/fisiologia , Emoções/fisiologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Córtex Pré-Frontal/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Feminino , GABAérgicos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , Glutamato Descarboxilase/metabolismo , Masculino , Transtornos Mentais/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Parvalbuminas/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , RNA Mensageiro/metabolismo , Caracteres Sexuais , Maturidade Sexual , Ácido Valproico/farmacologia
3.
J Neurosci ; 33(33): 13460-74, 2013 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-23946404

RESUMO

The decline of cognitive function has emerged as one of the greatest health threats of old age. Age-related cognitive decline is caused by an impacted neuronal circuitry, yet the molecular mechanisms responsible are unknown. C1q, the initiating protein of the classical complement cascade and powerful effector of the peripheral immune response, mediates synapse elimination in the developing CNS. Here we show that C1q protein levels dramatically increase in the normal aging mouse and human brain, by as much as 300-fold. This increase was predominantly localized in close proximity to synapses and occurred earliest and most dramatically in certain regions of the brain, including some but not all regions known to be selectively vulnerable in neurodegenerative diseases, i.e., the hippocampus, substantia nigra, and piriform cortex. C1q-deficient mice exhibited enhanced synaptic plasticity in the adult and reorganization of the circuitry in the aging hippocampal dentate gyrus. Moreover, aged C1q-deficient mice exhibited significantly less cognitive and memory decline in certain hippocampus-dependent behavior tests compared with their wild-type littermates. Unlike in the developing CNS, the complement cascade effector C3 was only present at very low levels in the adult and aging brain. In addition, the aging-dependent effect of C1q on the hippocampal circuitry was independent of C3 and unaccompanied by detectable synapse loss, providing evidence for a novel, complement- and synapse elimination-independent role for C1q in CNS aging.


Assuntos
Envelhecimento/metabolismo , Encéfalo/metabolismo , Complemento C1q/biossíntese , Animais , Comportamento Animal , Western Blotting , Encéfalo/fisiologia , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Imunoeletrônica
4.
Brain Res ; 1841: 149122, 2024 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-39009061

RESUMO

Women have a two-fold increased risk of developing Alzheimer's disease (AD) than men, yet the underlying mechanisms of this sex-specific vulnerability remain unknown. Here, we aimed at determining in the 5XFAD mouse model whether deficits in prefrontal-dependent cognitive functions, which are impacted in the preclinical stages of AD, appear earlier in females, and whether these cognitive deficits are associated with alterations in the activity of prefrontal parvalbumin (PV)-neurons that regulate prefrontal circuits activity. We observed that 3.5-month-old 5XFAD females, but not males, display impairments in spatial short-term recognition memory, a function that relies on the integrity of the prefrontal cortex. Hippocampal-dependent cognitive functions were intact in both sexes. We then observed that 5XFAD females have more prefrontal PV neurons expressing the marker of chronic activity FosB; this was inversely correlated with prefrontal-dependent cognitive performances. Our findings show for the first time sex-specific, early deregulation of prefrontal PV neurons activity, which is associated with early appearance of prefrontal-dependent cognitive functions in 5XFAD females providing a potential novel mechanism to the increased risk to AD in females.


Assuntos
Doença de Alzheimer , Modelos Animais de Doenças , Transtornos da Memória , Camundongos Transgênicos , Neurônios , Parvalbuminas , Córtex Pré-Frontal , Animais , Córtex Pré-Frontal/metabolismo , Parvalbuminas/metabolismo , Feminino , Neurônios/metabolismo , Masculino , Transtornos da Memória/fisiopatologia , Transtornos da Memória/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Memória Espacial/fisiologia , Camundongos , Reconhecimento Psicológico/fisiologia , Hipocampo/metabolismo , Caracteres Sexuais , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Humanos , Camundongos Endogâmicos C57BL
5.
Neuroscience ; 538: 68-79, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38157976

RESUMO

Inhibitory parvalbumin (PV) interneurons regulate the activity of neural circuits within brain regions involved in emotional processing, including the prefrontal cortex (PFC). Recently, rodent studies have implicated a stress-induced increase in prefrontal PV neuron activity in the development of anxiety behaviors, particularly in females. However, the mechanisms through which stress increases activity of prefrontal PV neurons remain unknown. The fast-spiking properties of PV neurons in part come from their expression of voltage-gated potassium (K+) ion channels, particularly Kv3.1 channels. We therefore suggest that stress-induced changes in Kv3.1 channels contribute to the appearance of an anxious phenotype following chronic stress in female mice. Here, we first showed that unpredictable chronic mild stress (UCMS) increased expression of Kv3.1 channels on prefrontal PV neurons in female mice, a potential mechanism underlying the previously observed hyperactivity of these neurons after stress. We then showed that female mice deficient in Kv3.1 channels displayed resilience to UCMS-induced anxiety-like behaviors. Altogether, our findings implicate Kv3.1 channels in the development of anxiety-like behaviors following UCMS, particularly in females, providing a novel mechanism to understand sex-specific vulnerabilities to stress-induced psychopathologies.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Masculino , Camundongos , Feminino , Animais , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Neurônios/metabolismo , Interneurônios/metabolismo , Encéfalo/metabolismo , Ansiedade/metabolismo , Canais de Potássio Shaw/metabolismo
6.
Psychoneuroendocrinology ; 148: 105997, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36470154

RESUMO

The increased vulnerability to stress-induced neuropsychiatric disorders in women, including anxiety disorders, does not emerge until pubertal onset, suggesting a role for ovarian hormones in organizing sex-specific vulnerability to anxiety. Parvalbumin (PV) interneurons in the prefrontal cortex are a potential target for these ovarian hormones. PV+ interneurons undergo maturation during the adolescent period and have been shown to be sensitive to stress and to mediate stress-induced anxiety in female mice. To test the idea that ovarian hormones at puberty are necessary for the acquisition of sensitivity to stress, hypothetically driving the response of PV+ interneurons to stress, we performed ovariectomy or sham surgery before pubertal onset in female mice. These mice then were exposed to four weeks of unpredictable chronic mild stress in adulthood. We then assessed anxiety-like behavior and PV/FosB colocalization in the medial PFC. Additionally, we assessed stress-induced anxiety-like behavior in female mice following ovariectomy in adulthood to determine if puberty is a sensitive period for ovarian hormones in mediating vulnerability to stress. We found that prepubertal ovariectomy protects against the development of anxiety-like behavior in adulthood, an effect not found following ovariectomy in adulthood. This effect may be independent of ovarian hormones on prefrontal PV+ interneurons response to stress.


Assuntos
Transtornos de Ansiedade , Ansiedade , Masculino , Humanos , Camundongos , Feminino , Animais , Ovariectomia , Interneurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Parvalbuminas/metabolismo , Hormônios
7.
eNeuro ; 10(3)2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36808099

RESUMO

Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.


Assuntos
Ansiedade , Parvalbuminas , Masculino , Camundongos , Feminino , Animais , Parvalbuminas/metabolismo , Ansiedade/patologia , Neurônios/metabolismo , Transtornos de Ansiedade , Emoções , Interneurônios/fisiologia , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/patologia
8.
Artigo em Inglês | MEDLINE | ID: mdl-38486965

RESUMO

The complexity of the classical inverted U-shaped relationship between cortisol levels and responses transposable to stress reactivity has led to an incomplete understanding of the mechanisms enabling healthy and toxic effects of stress on brain and behavior. A clearer, more detailed, picture of those relationships can be obtained by integrating cortisol effects on large-scale brain networks, in particular, by focusing on neural network configurations from the perspective of inhibition and excitation. A unifying view of Semon and Hebb's theories of cellular memory links the biophysical and metabolic changes in neuronal ensembles to the strengthening of collective synapses. In that sense, the neuronal capacity to record, store, and retrieve information directly relates to the adaptive capacity of its connectivity and metabolic reserves. Here, we use task-activated cell ensembles or simply engram cells as an example to demonstrate that the adaptive behavioral responses to stress result from collective synapse strength within and across networks of interneurons and excitatory ones.

9.
Neurosci Biobehav Rev ; 131: 1228-1242, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34718048

RESUMO

Stress is a major risk factor for neurodevelopmental and neuropsychiatric disorders, with the capacity to impact susceptibility to disease as well as long-term neurobiological and behavioral outcomes. Parvalbumin (PV) interneurons, the most prominent subtype of GABAergic interneurons in the cortex, are uniquely responsive to stress due to their protracted development throughout the highly plastic neonatal period and into puberty and adolescence. Additionally, PV + interneurons appear to respond to stress in a sex-specific manner. This review aims to discuss existing preclinical studies that support our overall hypothesis that the sex-and age-specific impacts of stress on PV + interneurons contribute to differences in individual vulnerability to stress across the lifespan, particularly in regard to sex differences in the diagnostic rate of neurodevelopmental and neuropsychiatric diseases in clinical populations. We also emphasize the importance of studying sex as a biological variable to fully understand the mechanistic and behavioral differences between males and females in models of neuropsychiatric disease.


Assuntos
Transtornos do Neurodesenvolvimento , Parvalbuminas , Animais , Feminino , Humanos , Recém-Nascido , Interneurônios , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parvalbuminas/farmacologia , Caracteres Sexuais
10.
Behav Brain Res ; 413: 113468, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34274375

RESUMO

Parvalbumin (PV)-expressing neurons have been implicated in the pathology of autism spectrum disorders (ASD). Loss of PV expression and/or reduced number of PV-expressing neurons have been reported not only in genetic and environmental rodent models of ASD, but also in post-mortem analyses of brain tissues from ASD vs. healthy control human subjects. PV-expressing neurons play a pivotal role in the maintenance of the balance between excitation and inhibition within neural circuits in part because of their fast-spiking properties. Their high firing rate is mostly regulated by the voltage-gated potassium channel Kv3.1. It is yet unknown whether disturbances in the electrophysiological properties of PV-expressing neurons per se can lead to behavioral disturbances. We assessed locomotor activity, social interaction, recognition and memory, and stereotypic behaviors in Kv3.1 wild-type (WT) and knockout (KO) mice. We then used Western Blot analyses to measure the impact of Kv3.1 deficiency on markers of GABA transmission (PV and GAD67) and neural circuit activity (Egr1). Deficiency in Kv3.1 channel is sufficient to induce social deficits, hyperactivity and stereotypic behaviors. These behavioral changes were independent of changes in GAD67 levels and associated with increased levels of PV protein in the prefrontal cortex and striatum. These findings reveal that a loss of PV expression is not a necessary factor to induce an ASD-like phenotype in mice and support the need for further investigation to fully understand the contribution of PV-expressing neurons to ASD pathology.


Assuntos
Transtorno do Espectro Autista , Comportamento Animal/fisiologia , Sintomas Comportamentais , Corpo Estriado , Parvalbuminas/metabolismo , Córtex Pré-Frontal , Agitação Psicomotora , Canais de Potássio Shaw/deficiência , Comportamento Social , Comportamento Estereotipado/fisiologia , Animais , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/fisiopatologia , Sintomas Comportamentais/metabolismo , Sintomas Comportamentais/fisiopatologia , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Agitação Psicomotora/metabolismo , Agitação Psicomotora/fisiopatologia
11.
Front Behav Neurosci ; 14: 581360, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33192367

RESUMO

Exposure to stress is recognized to be a triggering factor in several mood disorders, including depression and anxiety. There is very little understanding of why female subjects have a significantly higher risk for these conditions than males. Recent findings in male rodents indicated that prophylactic ketamine can prevent the development of a stress-induced depressive-like phenotype, providing a pharmacological tool to study the mechanisms underlying stress resilience. Unfortunately, none of these studies incorporated female subjects, nor did they provide a mechanistic understanding of the effects of ketamine on stress resilience. Our previous work identified the prefrontal glutamatergic and parvalbumin (PV) systems as potential molecular mechanisms underlying sex differences in susceptibility to stress-induced emotional deregulations. To further address this point, we treated male and female mice with a single dose of ketamine before exposure to a chronic stress paradigm to determine whether stress-resilience induced by a pre-exposure to ketamine is similar in males and females and whether modulation of the prefrontal glutamatergic and PV systems by ketamine is associated with these behavioral effects. Ketamine prevented chronic stress-induced changes in behaviors in males, which was associated with a reduction in expression of PV and the NMDA receptor NR1 subunit. Ketamine did not protect females against the effects of chronic stress and did not change significantly prefrontal gene expression. Our data highlight fundamental sex differences in the sustained effects of ketamine. They also further implicate prefrontal glutamatergic transmission and PV in resilience to chronic stress.

12.
Neurosci Biobehav Rev ; 105: 39-51, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31377218

RESUMO

Chronic stress-induced emotional disorders like anxiety and depression involve imbalances between the excitatory glutamatergic system and the inhibitory GABAergic system in the prefrontal cortex (PFC). However, the precise nature and trajectory of excitatory/inhibitory (E/I) imbalances in these conditions is not clear, with the literature reporting glutamatergic and GABAergic findings that are at times contradictory and inconclusive. Here we propose and discuss the hypothesis that chronic stress-induced emotional dysfunction involves hypoactivity of the PFC due to increased inhibition. We will also discuss E/I imbalances in the context of sex differences. In this review, we will synthesize research about how glutamatergic and GABAergic systems are perturbed by chronic stress and in related emotional disorders like anxiety and depression and propose ideas for reconciling contradictory findings in support of the hypothesis of over-inhibition. We will also discuss evidence for how aspects of the GABAergic system such as parvalbumin (PV) cells can be targeted therapeutically for reinstating activity and plasticity in the PFC and treating stress-related disorders.


Assuntos
Sintomas Afetivos , Ansiedade , Excitabilidade Cortical , Depressão , Inibição Neural , Córtex Pré-Frontal , Estresse Psicológico , Sintomas Afetivos/metabolismo , Sintomas Afetivos/fisiopatologia , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Excitabilidade Cortical/fisiologia , Depressão/metabolismo , Depressão/fisiopatologia , Humanos , Inibição Neural/fisiologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
13.
Transl Psychiatry ; 9(1): 99, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30792384

RESUMO

Dysfunction of prefrontal parvalbumin (PV+) interneurons has been linked with severe cognitive deficits as observed in several neurodevelopmental disorders including schizophrenia. However, whether a specific aspect of PV+ neurons deregulation, or a specific molecular mechanism within PV+ neurons is responsible for cognitive deficits and other behavioral impairments remain to be determined. Here, we induced cognitive deficits and altered the prefrontal PV system in mice by exposing them neonatally to the NMDA receptor antagonist ketamine. We observed that the cognitive deficits and hyperactivity induced by neonatal ketamine were associated with a downregulation of Npas4 expression specifically in PV+ neurons. To determine whether Npas4 downregulation-induced dysfunction of PV+ neurons could be a molecular contributor to the cognitive and behavioral impairments reported after neonatal ketamine, we used a transgenic Cre-Lox approach. Reduced Npas4 expression within PV+ neurons replicates deficits in short-term memory observed after neonatal ketamine, but does not reproduce disturbances in general activity. Our data show for the first time that the brain-specific transcription factor Npas4 may be an important contributor to PV+ neurons dysfunction in neurodevelopmental disorders, and thereby could contribute to the cognitive deficits observed in diseases characterized by abnormal functioning of PV+ neurons such as schizophrenia. These findings provide a potential novel therapeutic target to rescue the cognitive impairments of schizophrenia that remain to date unresponsive to treatments.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Disfunção Cognitiva/metabolismo , Interneurônios/metabolismo , Ketamina/farmacologia , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Esquizofrenia/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto , Memória de Curto Prazo/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/genética
14.
Acta Neuropathol Commun ; 7(1): 173, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31703603

RESUMO

Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.


Assuntos
Ansiedade/patologia , Consumo Excessivo de Bebidas Alcoólicas/patologia , Doenças Desmielinizantes/patologia , Etanol/toxicidade , Interneurônios/patologia , Fatores Etários , Animais , Ansiedade/induzido quimicamente , Ansiedade/psicologia , Consumo Excessivo de Bebidas Alcoólicas/psicologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/psicologia , Etanol/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Interneurônios/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia
15.
Sci Rep ; 9(1): 19772, 2019 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-31875035

RESUMO

Reduced activity of the prefrontal cortex (PFC) is seen in mood disorders including depression and anxiety. The mechanisms of this hypofrontality remain unclear. Because of their specific physiological properties, parvalbumin-expressing (PV+) inhibitory interneurons contribute to the overall activity of the PFC. Our recent work using a chronic stress mouse model showed that stress-induced increases in prefrontal PV expression correlates with increased anxiety-like behaviors in female mice. Our goal is now to provide a causal relationship between changes in prefrontal PV+ cells and changes in emotional behaviors in mice. We first show that, in addition to increasing overall level of PV expression, chronic stress increases the activity of prefrontal PV+ cells. We then used a chemogenetic approach to mimic the effects of chronic stress and specifically increase the activity of prefrontal PV+ cells. We observed that chemogenetic activation of PV+ cells caused an overall reduction in prefrontal activity, and that chronic activation of PV+ cells lead to increased anxiety-related behaviors in female mice only. These results demonstrate that activity of prefrontal PV+ cells could represent a novel sex-specific modulator of anxiety-related behaviors, potentially through changes in overall prefrontal activity. The findings also support the idea that prefrontal PV+ cells are worth further investigation to better understand mood disorders that are more prevalent in female populations.


Assuntos
Ansiedade/metabolismo , Comportamento Animal , Regulação da Expressão Gênica , Interneurônios/metabolismo , Parvalbuminas/biossíntese , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Animais , Ansiedade/patologia , Modelos Animais de Doenças , Feminino , Interneurônios/patologia , Masculino , Camundongos , Córtex Pré-Frontal/patologia , Estresse Psicológico/patologia
16.
Physiol Behav ; 93(1-2): 395-407, 2008 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-17961613

RESUMO

According to the maternal mediation hypothesis, brain and behavioural development in rodents is affected by environment-dependent variations in maternal care. Thus, it has been shown that early handling results in reduced behavioural and neuroendocrine responses to stressors and that these effects are associated with increased maternal care received during infancy. To investigate this further in mice, we chose a less artificial paradigm that is not confounded by human manipulation and reflects a more natural form of early environmental variation. We housed lactating C57BL/6 dams and their litters in cage systems composed of a nest cage (NC) and a foraging cage (FC) connected by a tunnel, and varied the dams' access to food by providing food either in the NC (NC dams) or FC (FC dams) until postnatal day 14. FC dams were more frequently observed in the FC than NC dams, and although the frequency of the dams being in physical contact with the pups did not differ between the two treatments, FC dams showed lower levels of active nursing than NC dams during the first week of lactation. These environment-dependent variations in maternal behaviour had sex-specific effects on the adult offspring's behavioural and HPA responses to stressors and altered their social behaviour in the home cage, with NC offspring showing higher levels of socio-positive behaviours than FC offspring. These results provide further independent evidence for the maternal mediation hypothesis and demonstrate that even subtle variations of the maternal environment can affect maternal care and induce persistent changes in offspring phenotype.


Assuntos
Corticosterona/sangue , Comportamento Materno/fisiologia , Comportamento Social , Meio Social , Estresse Psicológico/sangue , Adaptação Fisiológica , Adaptação Psicológica , Animais , Meio Ambiente , Comportamento Exploratório/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais , Estatísticas não Paramétricas , Estresse Psicológico/psicologia
17.
Physiol Behav ; 94(3): 393-404, 2008 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-18346766

RESUMO

Recent studies in rats and mice suggest that developmental plasticity of HPA-stress and fear responses could be mediated by environment-dependent variations in maternal behaviour. The present study was designed to examine this question further by varying the adversity of the maternal environment to study its effects on nest-attendance and maternal care and on the HPA and fear responses in the adult offspring. C57BL/6 dams and their litter were housed in a cage system composed of a nest cage (NC) and a foraging cage (FC) connected by a tunnel. Using a 2 x 2 factorial design, we varied the maternal foraging environment (FC) by the presence or absence of rat odour (feces) and shelters (MouseHouse and tube) from postnatal days 1-14 and assessed the adult offspring's corticosterone response to isolation/novelty stress and their behaviour in three tests of fearfulness (elevated-O-maze, open-field, free exploration). While the presence of shelters in the FC reduced time spent in the NC (nest site attendance), the presence of rat odour in the FC increased active maternal care without altering nest site attendance. Alterations of the offspring's HPA and fear responses were rather subtle. The presence of shelters in the dam's foraging environment decreased fearfulness in the offspring in the free exploration test. In addition, males reared by dams exposed to rat odour were less fearful in the open-field test, and both males and females reared by dams without shelters and rat odour in the FC showed a greater corticosterone response to isolation/novelty stress. Multiple regression analysis indicated a negative relationship between maternal licking/grooming and fearfulness in males and a positive relationship between nest site attendance and fearfulness in females. Taken together, these results indicate that mouse dams adjust specific aspects of maternal behaviour in response to the specific properties of their environment, and that active maternal care and nest site attendance are two aspects of maternal behaviour that may affect the offspring's stress and fear systems independent of each other and in a sex-specific way.


Assuntos
Animais Recém-Nascidos/fisiologia , Medo , Comportamento Materno/fisiologia , Odorantes , Meio Social , Estresse Psicológico/fisiopatologia , Fatores Etários , Análise de Variância , Animais , Comportamento Animal , Tamanho Corporal , Peso Corporal , Corticosterona/sangue , Comportamento Exploratório/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Ratos , Fatores Sexuais
18.
Mol Neurobiol ; 55(3): 2591-2602, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28421533

RESUMO

The prefrontal cortex (PFC) is highly sensitive to the effects of stress, a known risk factor of mood disorders including anxiety and depression. Abnormalities in PFC functioning have been well described in humans displaying stress-induced depressive symptoms, and hypoactivity of the PFC is now recognized to be a key feature of the depressed brain. However, little is known about the causes and mechanisms leading to this altered prefrontal functional activity in the context of stress-related mood disorders. We previously showed that unpredictable chronic mild stress (UCMS) in mice increases prefrontal expression of parvalbumin (PV), an activity-dependent calcium-binding albumin protein expressed in a specific subtype of GABAergic neurons, highlighting a potential mechanism through which chronic stress leads to hypofunction of the PFC. In this study, we aimed to investigate the mechanisms by which chronic stress alters the prefrontal GABA system. We hypothesized that chronic stress-induced enhancement of glutamatergic transmission in the PFC is a crucial contributing factor to changes within the prefrontal GABAergic and, specifically, PV system. BALB/c male and female mice were exposed to daily handling (control) or 2 or 4 weeks of UCMS. Female mice displayed a more severe altered phenotype than males, as shown by increased anxiety- and depressive-like behaviors and deficits in PFC-dependent cognitive abilities, particularly after exposure to 2 weeks of UCMS. This behavioral phenotype was paralleled by a large increase in prefrontal PV messenger RNA (mRNA) and number of PV-expressing neurons, supporting our previous findings. We further showed that the expression of pre- and postsynaptic markers of glutamatergic transmission (VGlut1 presynaptic terminals and pERK1/2, respectively) onto PV neurons was increased by 2 weeks of UCMS in a sex-specific manner; this was associated with sex-specific changes in the mRNA expression of the NR2B subunit of the NMDA receptor. These findings provide evidence of increased glutamatergic transmission onto prefrontal PV neurons, particularly in female mice, which could potentially contribute to their increased PV expression and the extent of their behavioral impairment following UCMS. Finally, our analysis of activity of subcortical regions sending glutamatergic afferents to the PFC reveals that glutamatergic neurons from the basolateral amygdala might be specifically involved in UCMS-induced changes in prefrontal glutamatergic transmission.


Assuntos
Ácido Glutâmico/metabolismo , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Estresse Psicológico/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Animais , Doença Crônica , Feminino , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Córtex Pré-Frontal/patologia , Estresse Psicológico/patologia , Estresse Psicológico/psicologia
19.
Neuroscience ; 390: 265-277, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30179643

RESUMO

Adolescence is a window of vulnerability to environmental factors such as chronic stress that can disrupt brain development and cause long-lasting behavioral dysfunction, as seen in disorders like depression, anxiety, and schizophrenia. There are also sex differences in the prevalence of these disorders across the lifespan. However, the mechanisms of how adolescent stress contributes to neuropsychiatric phenotypes are not well understood, nor are the mediating effects of sex. We hypothesize that adolescent stress disrupts the γ-aminobutyric acid (GABA) system in the prefrontal cortex (PFC) in a sex-specific manner, as this system matures during adolescence and plays an important role in cognitive and emotional functioning. We exposed male and female mice to unpredictable chronic mild stress (UCMS) during adolescence (post-natal day [PND] 28-42). One cohort underwent testing for PFC-related behavioral and molecular changes 24 h following the cessation of stress (late adolescence); a separate cohort was tested approximately 2.5 weeks after the end of UCMS (adulthood). We observed an age-related decline in anxiety-like behaviors in control mice, while mice stressed in adolescence showed elevated anxiety-like behaviors in both adolescence and adulthood. PFC-dependent cognitive functioning was also impaired in adult males stressed in adolescence. Adolescent stress disrupted expression patterns of parvalbumin (PV) and perineuronal nets (PNNs) in the PFC, as well as NMDA receptor subunit composition, in a sex- and age-specific manner. The findings presented here contribute to understanding how adolescent stress may lead to neuropsychiatric disorders such as anxiety by disrupting the development of the PFC and emotional behaviors.


Assuntos
Ansiedade/fisiopatologia , Córtex Pré-Frontal/crescimento & desenvolvimento , Caracteres Sexuais , Estresse Psicológico/fisiopatologia , Animais , Depressão , Feminino , Neurônios GABAérgicos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Parvalbuminas/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
20.
Dev Cogn Neurosci ; 28: 29-40, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29102727

RESUMO

Executive functions (EF) reached full maturity during the transition from adolescence to adulthood. Human studies provide important information about adolescent developmental trajectories; however, little remains known about the neural circuits underlying the acquisition of mature EF. Ethical and technical considerations with human subjects limit opportunities to design experimental studies that allows for an in-depth understanding of developmental changes in neural circuits that regulate cognitive maturation. Preclinical models can offer solutions to this problem. Unfortunately, current rodent models of adolescent development have inherent flaws that limit their translational value. For instance, females are often omitted from studies, preventing the assessment of potential sex-specific developmental trajectories. Furthermore, it remains unclear whether cognitive developmental changes in rodents are similar to those observed in humans. Here, we tested adolescent and adult male and female mice in a neurocognitive battery of assays. Based on this approach, we assessed mice performances within distinct subdomains of EF, and observed similarities with human developmental trajectories. Furthermore, the sex-specific cognitive changes we observed were paralleled by molecular and neural activity changes demonstrating that our approach can be used in future research to assess the contribution of precise neural circuits to adolescent cognitive maturation.


Assuntos
Cognição/fisiologia , Função Executiva/fisiologia , Adolescente , Adulto , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA