Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Neuropharmacol ; 22(5): 935-962, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37403395

RESUMO

Exposure to acute and chronic stress has a broad range of structural effects on the brain. The brain areas commonly targeted in the stress response models include the hippocampus, the amygdala, and the prefrontal cortex. Studies in patients suffering from the so-called stress-related disorders -embracing post-traumatic stress, major depressive and anxiety disorders- have fairly replicated animal models of stress response -particularly the neuroendocrine and the inflammatory models- by finding alterations in different brain areas, even in the early neurodevelopment. Therefore, this narrative review aims to provide an overview of structural neuroimaging findings and to discuss how these studies have contributed to our knowledge of variability in response to stress and the ulterior development of stress-related disorders. There are a gross number of studies available but neuroimaging research of stress-related disorders as a single category is still in its infancy. Although the available studies point at particular brain circuitries involved in stress and emotion regulation, the pathophysiology of these abnormalities -involving genetics, epigenetics and molecular pathways-, their relation to intraindividual stress responses -including personality characteristics, self-perception of stress conditions…-, and their potential involvement as biomarkers in diagnosis, treatment prescription and prognosis are discussed.


Assuntos
Transtorno Depressivo Maior , Transtornos de Estresse Pós-Traumáticos , Animais , Humanos , Encéfalo/diagnóstico por imagem , Transtornos de Ansiedade , Biomarcadores , Imageamento por Ressonância Magnética
2.
Curr Top Behav Neurosci ; 64: 105-132, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37528309

RESUMO

Fear extinction memories are strongly modulated by sex and hormonal status, but the exact mechanisms are still being discovered. In humans, there are some basal and task-related features in which male and female individuals differ in fear conditioning paradigms. However, analyses considering the effects of sex hormones demonstrate a role for estradiol in fear extinction memory consolidation. Translational studies are taking advantage of the convergent findings between species to understand the brain structures implicated. Nevertheless, the human brain is complex and the transfer of these findings into the clinics remains a challenge. The promising advances in the field together with the standardization of fear extinction methodologies in humans will benefit the design of new personalized therapies.


Assuntos
Extinção Psicológica , Medo , Feminino , Masculino , Humanos , Caracteres Sexuais , Hormônios Esteroides Gonadais/farmacologia , Estradiol/farmacologia
3.
Neurosci Biobehav Rev ; 151: 105224, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37156310

RESUMO

Restriction of free movement has historically been used as a model for inducing acute and chronic stress in laboratory animals. This paradigm is one of the most widely employed experimental procedures for basic research studies of stress-related disorders. It is easy to implement, and it rarely involves any physical harm to the animal. Many different methods have been developed with variations in the apparatuses used and the degree of limitation of movement. Unfortunately, very few studies directly compare the differential impact of the distinct protocols. Additionally, restraint and immobilization terms are not differentiated and are sometimes used interchangeably in the literature. This review offers evidence of great physiological differences in the impact of distinct restraint and immobilization procedures in rats and mice and emphasizes the need for a standardized language on this topic. Moreover, it illustrates the necessity of additional systematic studies that compare the effects of the distinct methodologies, which would help to decide better which procedure should be used depending on the objectives of each particular study.


Assuntos
Restrição Física , Roedores , Ratos , Camundongos , Animais , Restrição Física/métodos , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal/fisiologia , Corticosterona , Estresse Psicológico , Estresse Fisiológico
4.
bioRxiv ; 2023 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-37205515

RESUMO

Combining the use of ex vivo and in vivo optogenetics, viral tracing, electrophysiology and behavioral testing, we show that the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) gates anxiety-controlling circuits by differentially affecting synaptic efficacy at projections from the basolateral amygdala (BLA) to two different subdivisions of the dorsal subdivision of the bed nucleus of the stria terminalis (BNST), modifying the signal flow in BLA-ovBNST-adBNST circuits in such a way that adBNST is inhibited. Inhibition of adBNST is translated into the reduced firing probability of adBNST neurons during afferent activation, explaining the anxiety-triggering actions of PACAP in BNST, as inhibition of adBNST is anxiogenic. Our results reveal how innate, fear-related behavioral mechanisms may be controlled by neuropeptides, PACAP specifically, at the level of underlying neural circuits by inducing long-lasting plastic changes in functional interactions between their different structural components.

5.
Neuropharmacology ; 224: 109344, 2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36402246

RESUMO

Posttraumatic stress disorder (PTSD) is a highly disabling psychiatric condition that may arise after exposure to acute and severe trauma. It is a highly prevalent mental disorder worldwide, and the current treatment options for these patients remain limited due to low effectiveness. The time window right after traumatic events provides clinicians with a unique opportunity for preventive interventions against potential deleterious alterations in brain function that lead to PTSD. Some studies pointed out that PTSD patients present an abnormal function of the hypothalamic-pituitary-adrenal axis that may contribute to a vulnerability toward PTSD. Moreover, glucocorticoids have arisen as a promising option for preventing the disorder's development when administered in the aftermath of trauma. The present work compiles the recent findings of glucocorticoid administration for the prevention of a PTSD phenotype, from human studies to animal models of PTSD. Overall, glucocorticoid-based therapies for preventing PTSD demonstrated moderate evidence in terms of efficacy in both clinical and preclinical studies. Although clinical studies point out that glucocorticoids may not be effective for all patients' subpopulations, those with adequate traits might greatly benefit from them. Preclinical studies provide precise insight into the mechanisms mediating this preventive effect, showing glucocorticoid-based prevention to reduce long-lasting behavioral and neurobiological abnormalities caused by traumatic stress. However, further research is needed to delineate the precise mechanisms and the extent to which these interventions can translate into lower PTSD rates and morbidity. This article is part of the Special Issue on 'Fear, Anxiety and PTSD'.


Assuntos
Glucocorticoides , Transtornos de Estresse Pós-Traumáticos , Animais , Humanos , Glucocorticoides/uso terapêutico , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transtornos de Estresse Pós-Traumáticos/prevenção & controle , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Ansiedade , Hidrocortisona
6.
Neurobiol Stress ; 18: 100448, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35685680

RESUMO

Background: Pituitary adenylate cyclase-activating polypeptide (PACAP) receptor gene polymorphism has been postulated as a potential sex-specific diagnostic biomarker of trauma-related disorders. However, no research to date has evaluated whether the PACAPergic system may act as a vulnerability/resilience neuromechanism to trauma-induced psychopathology in healthy participants without heightened risk to experience traumatic events. Methods: Here, we compared the amygdala and hippocampus response to fearful faces in participants with at-risk genotype versus non-risk participants from the Human Connectome Project (n = 991; 53.4% female). Results: Increased hippocampal response to fearful faces in the female risk group emerged in sex by genetic risk interaction. Conclusions: Our findings revealed the first sex-specific neurogenetic vulnerability factor to trauma-related disorders, and emphasize the importance of prevention-based strategies to ameliorate neuropsychiatric pathophysiology.

7.
Biol Sex Differ ; 13(1): 28, 2022 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-35690790

RESUMO

BACKGROUND: Memory consolidation is a process required for the formation of long-term memories. The G-protein-coupled receptor (GPCR) neurokinin-3-receptor (Nk3R) and its interactions with sex hormones seem important for the modulation of fear memory consolidation: Nk3R antagonism in male mice impairs fear memory, but enhances it in females. However, the involvement of the Nk3R as a modulator of other memories in both sexes remains unexplored. METHODS: We use the novel object recognition paradigm to test the effect of a systemic blockade of Nk3R during memory consolidation. Further, we assess the expression of estrogen receptor α, estrogen receptor ß, and androgen receptor and heterodimerization with Nk3R in the medial prefrontal cortex (mPFC) and dorsal hippocampus (DH) of mice. RESULTS: Nk3R systemic antagonism elicited decreased memory consolidation in males while it enhanced it in females during proestrus. Nk3R analysis in the different subregions of the mPFC and the DH showed a higher expression in males than females. Moreover, females presented upregulation of the androgen receptor in the CA1 and the estrogen receptor beta in the cingulate cortex, CA1, and dentate gyrus. Overall, males presented an upregulation of the estrogen receptor alpha. We also explored the heterodimerization of GCPR membrane sex hormone receptors with the Nk3R. We found a higher percentage of Nk3R-membrane G-protein estrogen receptors heterodimers in the prelimbic cortex of the mPFC in females, suggesting an interaction of estradiol with Nk3R in memory consolidation. However, males presented a higher percentage of Nk3R-membrane G-protein androgen receptors heterodimers compared to females, pointing to an interaction of testosterone with Nk3R in memory consolidation. CONCLUSION: These data propose novel ideas on functional interactions between Nk3R, sex hormones, estrogen receptors, and androgen receptors in memory consolidation.


Assuntos
Consolidação da Memória , Receptores Androgênicos , Receptores da Neurocinina-3/metabolismo , Animais , Receptor beta de Estrogênio/metabolismo , Feminino , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/farmacologia , Masculino , Consolidação da Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo
8.
Elife ; 102021 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-34787081

RESUMO

De novo protein synthesis is required for synapse modifications underlying stable memory encoding. Yet neurons are highly compartmentalized cells and how protein synthesis can be regulated at the synapse level is unknown. Here, we characterize neuronal signaling complexes formed by the postsynaptic scaffold GIT1, the mechanistic target of rapamycin (mTOR) kinase, and Raptor that couple synaptic stimuli to mTOR-dependent protein synthesis; and identify NMDA receptors containing GluN3A subunits as key negative regulators of GIT1 binding to mTOR. Disruption of GIT1/mTOR complexes by enhancing GluN3A expression or silencing GIT1 inhibits synaptic mTOR activation and restricts the mTOR-dependent translation of specific activity-regulated mRNAs. Conversely, GluN3A removal enables complex formation, potentiates mTOR-dependent protein synthesis, and facilitates the consolidation of associative and spatial memories in mice. The memory enhancement becomes evident with light or spaced training, can be achieved by selectively deleting GluN3A from excitatory neurons during adulthood, and does not compromise other aspects of cognition such as memory flexibility or extinction. Our findings provide mechanistic insight into synaptic translational control and reveal a potentially selective target for cognitive enhancement.


Assuntos
Memória/fisiologia , Biossíntese de Proteínas/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais
9.
Curr Protoc ; 1(5): e102, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33950571

RESUMO

Fear conditioning (FC) is a widely accepted tool for the assessment of learning and memory processes in rodents related to normal and dysregulated acquired fear. The study of sex differences in fear learning and memory is vast and currently increasing. Sex hormones have proven to be crucial for fear memory formation in males and females, and several methods have been developed to assess this hormonal state in rats and mice. Herein, we explain a routine FC and extinction protocol, together with the evaluation of sex hormonal state in male and female rodents. We explain three protocols for the evaluation of this hormonal state directly from blood samples extracted during the procedure or indirectly through histological verification of the estrous cycle for females or behavioral assessment of social hierarchies in males. Although females have typically been considered to present great variability in sex hormones, it is highlighted that sex hormone assessment in males is as variable as in females and equally important for fear memory formation. The readout of these protocols has had a great impact on different fields of fear learning and memory study and appears essential when studying FC. The proven interaction with drugs involved in the modulation of these processes makes sex hormone assessment during FC a valuable tool for the development of effective treatments for fear-related disorders in men and women. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Fear conditioning and fear extinction Basic Protocol 2: Blood collection for direct measurement of sex hormone levels in fear conditioning Basic Protocol 3: Indirect measurement of sex hormones in females during fear conditioning Basic Protocol 4: Assessment of dominance status in males before a fear conditioning protocol Support Protocol: Construction of a confrontation tube.


Assuntos
Extinção Psicológica , Medo , Animais , Feminino , Hormônios Esteroides Gonadais , Masculino , Camundongos , Ratos , Roedores , Caracteres Sexuais
10.
J Neurosci ; 41(5): 901-910, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33472824

RESUMO

Post-traumatic stress disorder (PTSD) is characterized by hypervigilance, increased reactivity to unpredictable versus predictable threat signals, deficits in fear extinction, and an inability to discriminate between threat and safety. First-line pharmacotherapies for psychiatric disorders have limited therapeutic efficacy in PTSD. However, recent studies have advanced our understanding of the roles of several limbic neuropeptides in the regulation of defensive behaviors and in the neural processes that are disrupted in PTSD. For example, preclinical studies have shown that blockers of tachykinin pathways, such as the Tac2 pathway, attenuate fear memory consolidation in mice and thus might have unique potential as early post-trauma interventions to prevent PTSD development. Targeting this pathway might also be beneficial in regulating other symptoms of PTSD, including trauma-induced aggressive behavior. In addition, preclinical and clinical studies have shown the important role of angiotensin receptors in fear extinction and the promise of using angiotensin II receptor blockade to reduce PTSD symptom severity. Additional preclinical studies have demonstrated that the oxytocin receptors foster accurate fear discrimination by facilitating fear responses to predictable versus unpredictable threats. Complementary human imaging studies demonstrate unique neural targets of intranasal oxytocin and compare its efficacy with well-established anxiolytic treatments. Finally, promising data from human subjects have demonstrated that a selective vasopressin 1A receptor antagonist reduces anxiety induced by unpredictable threats. This review highlights these novel promising targets for the treatment of unique core elements of PTSD pathophysiology.


Assuntos
Ansiedade/metabolismo , Emoções/fisiologia , Sistema Límbico/metabolismo , Neuropeptídeos/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Animais , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Emoções/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Humanos , Sistema Límbico/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/metabolismo , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , Receptores de Taquicininas/antagonistas & inibidores , Receptores de Taquicininas/metabolismo , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transtornos de Estresse Pós-Traumáticos/psicologia , Taquicininas/antagonistas & inibidores , Taquicininas/metabolismo
11.
Nat Commun ; 11(1): 782, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32034128

RESUMO

Food addiction is linked to obesity and eating disorders and is characterized by a loss of behavioral control and compulsive food intake. Here, using a food addiction mouse model, we report that the lack of cannabinoid type-1 receptor in dorsal telencephalic glutamatergic neurons prevents the development of food addiction-like behavior, which is associated with enhanced synaptic excitatory transmission in the medial prefrontal cortex (mPFC) and in the nucleus accumbens (NAc). In contrast, chemogenetic inhibition of neuronal activity in the mPFC-NAc pathway induces compulsive food seeking. Transcriptomic analysis and genetic manipulation identified that increased dopamine D2 receptor expression in the mPFC-NAc pathway promotes the addiction-like phenotype. Our study unravels a new neurobiological mechanism underlying resilience and vulnerability to the development of food addiction, which could pave the way towards novel and efficient interventions for this disorder.


Assuntos
Dependência de Alimentos/fisiopatologia , Núcleo Accumbens/fisiologia , Córtex Pré-Frontal/fisiologia , Receptores de Dopamina D2/genética , Animais , Modelos Animais de Doenças , Comportamento Alimentar/fisiologia , Dependência de Alimentos/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Camundongos Knockout , Vias Neurais/fisiologia , Receptor CB1 de Canabinoide/genética , Transmissão Sináptica , Regulação para Cima
12.
Genes Brain Behav ; 19(3): e12621, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31661603

RESUMO

Neuropeptide S (NPS) is a neuropeptide involved in the regulation of fear. Because safety learning is impaired in patients suffering from anxiety-related psychiatric disorders, and polymorphisms of the human neuropeptide S receptor (NPSR) gene have also been associated with anxiety disorders, we wanted to investigate whether NPSR-deficiency interferes with safety learning, and how prior stress would affect this type of learning. We first investigated the effect of pre-exposure to two different types of stressors (electric stimuli or immobilization) on safety learning in female and male C57Bl/6 mice, and found that while stress induced by electric stimuli enhanced safety learning in males, there were no differences in safety learning following immobilization stress. To further investigate the role of the NPS system in stress-induced modulation of safety learning, we exposed NPSR-deficient mice to stress induced by electric stimuli 10 days before safety learning. In nonstressed male mice, NPSR-deficiency enhanced safety learning. As in male C57Bl/6 mice, pre-exposure to electric stimuli increased safety learning in male NPSR +/+ mice. This pre-exposure effect was blocked in NPSR-deficient male mice showing impaired, but still intact, safety learning in comparison to their NPSR +/+ and NPSR +/- littermates. There was neither a pre-exposure nor a genotype effect in female mice. Our findings provide evidence that pre-exposure to stress induced by electric stimuli enhances safety learning in male mice, and that NPSR-deficiency prevents the beneficial effect of stress exposure on safety learning. We propose an inverted U-shape relationship between stress and safety learning.


Assuntos
Condicionamento Clássico , Receptores de Neuropeptídeos/genética , Animais , Estimulação Elétrica , Medo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Neuropeptídeos/deficiência , Fatores Sexuais
13.
Neurosci Biobehav Rev ; 107: 154-165, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31520677

RESUMO

Approximately one third of individuals who experience a severe traumatic event will develop posttraumatic stress disorder (PTSD). It is crucial to identify what factors may be associated with increased or decreased risk for PTSD. We conducted an umbrella review of systematic reviews and meta-analyses of risk/protective factors for PTSD and assessed and graded the evidence of the association between each factor and PTSD. Thirty-three systematic reviews and meta-analyses were included and 130 potential risk factors were identified. Of those, 57 showed a significant association with PTSD. Being female or being indigenous people of the Americas, among the sociodemographic factors; history of physical disease and family history of psychiatric disorder, among the pretrauma factors; and cumulative exposure to potentially traumatic experiences, trauma severity, and being trapped during an earthquake, among the peritrauma factors, showed convincing or highly suggestive evidence of an association with PTSD. Data from prospective studies were less conclusive. Our results have the potential of helping refine PTSD prediction models and contributing to the design of prevention strategies.


Assuntos
Transtornos de Estresse Pós-Traumáticos/etiologia , Humanos , Metanálise como Assunto , Literatura de Revisão como Assunto , Fatores de Risco , Transtornos de Estresse Pós-Traumáticos/psicologia
14.
Neuropharmacology ; 144: 345-357, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30439419

RESUMO

Δ9-tetrahydrocannabinol (THC) consumption during adolescence is reported to be a risk factor for the appearance of psychiatric disorders later in life. The interaction between genetic or environmental events and cannabinoid exposure in the adolescent period can also contribute to exacerbate behavioural deficits in adulthood. Here we investigate the effects of THC treatment as well as the consequences of concomitant THC and stress exposure during adolescence in the extinction of fear memory in adult mice. Adolescent mice treated with THC and exposed to stress exhibit impaired cued fear extinction in adulthood. However, no effect was observed in animals exposed to these two factors separately. Notably, resistance to fear extinction was associated with decreased neuronal activity in the basolateral amygdala (BLA) and the infralimbic prefrontal cortex, suggesting a long-term dysregulation of the fear circuit. These changes in neuronal activation were paralleled with structural plasticity alterations. Indeed, an increase of immature dendritic spines in pyramidal neurons of the BLA was revealed in mice simultaneously exposed to THC and stress. Corticosterone levels were also enhanced after the cued fear conditioning session in the same experimental group. These results show that an interaction between cannabis exposure and stress during adolescence may lead to long-term anxiety disorders characterized by the presence of pathological fear.


Assuntos
Encéfalo/crescimento & desenvolvimento , Dronabinol/efeitos adversos , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Psicotrópicos/efeitos adversos , Estresse Psicológico/fisiopatologia , Animais , Ansiedade/etiologia , Ansiedade/patologia , Ansiedade/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Encéfalo/fisiopatologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Espinhas Dendríticas/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Medo/psicologia , Feminino , Masculino , Uso da Maconha/metabolismo , Uso da Maconha/patologia , Uso da Maconha/psicologia , Camundongos Endogâmicos C57BL , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Células Piramidais/fisiologia , Maturidade Sexual , Estresse Psicológico/patologia
15.
Mol Psychiatry ; 23(11): 2122-2132, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29298989

RESUMO

We address some of the current limitations of translational research in fear memory and suggest alternatives that might help to overcome them. Appropriate fear responses are adaptive, but disruption of healthy fear memory circuits can lead to anxiety and fear-based disorders. Stress is one of the main environmental factors that can disrupt memory circuits and constitutes as a key factor in the etiopathology of these psychiatric conditions. Current therapies for anxiety and fear-based disorders have limited success rate, revealing a clear need for an improved understanding of their neurobiological basis. Although animal models are excellent for dissecting fear memory circuits and have driven tremendous advances in the field, translation of these findings into the clinic has been limited so far. Animal models of stress-induced pathological fear combined with powerful cutting-edge techniques would help to improve the translational value of preclinical studies. We also encourage combining animal and human research, including psychiatric patients in order to find new pharmacological targets with real therapeutic potential that will improve the extrapolation of the findings. Finally, we highlight novel neuroimaging approaches that improve our understanding of anxiety and fear-based disorders.


Assuntos
Medo/fisiologia , Memória/fisiologia , Pesquisa Translacional Biomédica/tendências , Animais , Ansiedade/terapia , Transtornos de Ansiedade/psicologia , Extinção Psicológica/fisiologia , Medo/psicologia , Humanos , Modelos Animais , Estresse Psicológico/fisiopatologia , Pesquisa Translacional Biomédica/métodos
16.
Front Psychiatry ; 9: 778, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30705647

RESUMO

Stress and trauma profoundly influence psychiatric biobehavioral outcomes. The identification of treatment and biomarker targets would be accelerated by a broad understanding of the biological responses to these events. The goal of this study was to determine genes responsive to auditory fear conditioning (FC), a well-characterized amygdala-dependent rodent model of threat-exposure, in the presence or absence of prior stress history, providing insight into the physiological processes underlying response to trauma. RNA-sequencing was performed in blood and amygdala from mice that underwent fear conditioning with (Immo+FC) and without (FC) prior immobilization stress, a paradigm that induces HPA axis, and behavioral stress sensitization. In the amygdala, 607 genes were regulated by FC vs. home-cage (HC) controls, and 516 genes differed in stress-sensitized mice (Immo+FC vs. FC). In the former, we observed an enhancement of specific biological processes involved in learning and synaptic transmission, and in the latter processes associated with cell proliferation and the cellular response to drugs. In the blood of stress-sensitized animals, 468 genes were dynamically regulated when compared to FC, and were enriched for the biological pathways of inflammation and cytokine signaling. This study identified genes and pathways that respond to threat in the amygdala and blood of mice with and without a prior stress history and reveals the impact of stress history on subsequent inflammation. Future studies will be needed to examine the role of these dynamically regulated genes may play in human clinical stress and trauma-related disorders.

17.
Biol Psychiatry ; 83(3): 284-295, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29054677

RESUMO

BACKGROUND: Molecular mechanisms underlying psychological sequelae of exposure to stressful experiences, such as posttraumatic stress disorder (PTSD) and depression, are not well understood. METHODS: Using convergent evidence from animal and human transcriptomic and genomic studies, we aimed to identify genetic mechanisms underlying depression and anxiety after traumatic experiences. RESULTS: From a transcriptome-wide analysis in mice, we found the Ppm1f gene to be differentially expressed in the amygdala and medial prefrontal cortex (mPFC) a week after immobilization stress. Next, we found that PPM1F messenger RNA levels in human blood were downregulated in cases with symptoms of comorbid PTSD and depression and consistently in cases with anxiety symptoms in a separate human dataset. Furthermore, we showed that a genetic variant of PPM1F, rs17759843, was associated with comorbid PTSD and depression and with PPM1F expression in both human brain and blood. Given prior reported mechanistic links between PPM1F and CAMK2 (CAMKII), we examined blood messenger RNA level of CAMK2G in humans and found it to be lower in cases with comorbid PTSD and depression. We also found that PPM1F protein levels and colocalization with CAMK2G were altered in amygdala and mPFC of male mice. Additionally, we found that a systemic dose of corticosterone blocked the depressive-like phenotype elicited by stress in female mice. Lastly, corticosterone rescued the anxiety-like phenotype and messenger RNA levels of Ppm1f in amygdala and mPFC in male mice and in mPFC of female mice. CONCLUSIONS: Taken together, our data suggest a mechanistic pathway involving PPM1F and CAMK2G in stress- and trauma-related manifestation of anxiety and depression across species.


Assuntos
Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Transtorno Depressivo/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Córtex Pré-Frontal/metabolismo , Transtornos de Estresse Pós-Traumáticos/metabolismo , Estresse Psicológico/metabolismo , Animais , Ansiedade/epidemiologia , Comportamento Animal/fisiologia , Comorbidade , Transtorno Depressivo/epidemiologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Estresse Pós-Traumáticos/epidemiologia , Estresse Psicológico/epidemiologia , Transcriptoma/genética
18.
Cereb Cortex ; 28(4): 1233-1244, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28203747

RESUMO

Despite extensive research on the impact of emotional stressors on brain function using immediate-early genes (e.g., c-fos), there are still important questions that remain unanswered such as the reason for the progressive decline of c-fos expression in response to prolonged stress and the neuronal populations activated by different stressors. This study tackles these 2 questions by evaluating c-fos expression in response to 2 different emotional stressors applied sequentially, and performing a fluorescent double labeling of c-Fos protein and c-fos mRNA on stress-related brain areas. Results were complemented with the assessment of the hypothalamic-pituitary-adrenal axis activation. We showed that the progressive decline of c-fos expression could be related to 2 differing mechanisms involving either transcriptional repression or changes in stimulatory inputs. Moreover, the neuronal populations that respond to the different stressors appear to be predominantly separated in high-level processing areas (e.g., medial prefrontal cortex). However, in low-hierarchy areas (e.g., paraventricular nucleus of the hypothalamus) neuronal populations appear to respond unspecifically. The data suggest that the distinct physiological and behavioral consequences of emotional stressors, and their implication in the development of psychopathologies, are likely to be closely associated with neuronal populations specifically activated by each stressor.


Assuntos
Encéfalo/citologia , Regulação da Expressão Gênica/fisiologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Fisiológico/fisiologia , Hormônio Adrenocorticotrópico/sangue , Animais , Autorradiografia , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Masculino , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
19.
Neuropharmacology ; 116: 188-195, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28025095

RESUMO

Deficits in fear extinction learning are hypothesized to underlie the development of posttraumatic stress disorder (PTSD). Such deficits may, in part, be due to genetic and epigenetic variation in the stress related gene FKBP5. Conversely, altering FKBP5 epigenetic responses during memory consolidation may rescue extinction deficits making it a target for acute intervention to prevent the development of PTSD. Study 1 (Humans) examines if FKBP5 single nucleotide polymorphisms (SNPs) and PTSD symptom domains (re-experiencing, avoidance/numbing, hyperarousal) are associated with abnormal fear extinction phenotypes identified using latent growth mixture modeling (LGMM). Study 2 (Mice) tests if increasing doses of dexamethasone administered prior to extinction alters Fkbp5 mRNA production in the amygdala after extinction and recall and prevents the development of abnormal extinction phenotypes. In humans, abnormal extinction was associated with the TT homozygous genotype of FKBP5 SNPs RS9470080 and RS1360780, and hyperarousal symptoms. In mice, dexamethasone 300 µg/kg was associated with increased amygdala Fkbp5 mRNA following extinction and robust extinction learning while lower doses were not associated with amygdala Fkbp5 mRNA or differences in extinction learning. Further, mice that extinguished on dexamethasone 300 µg/kg maintained low levels of freezing behavior during recall training while mRNA levels were no longer elevated. Together, findings indicate that FKBP5 confers risk for fear extinction deficits. However, this risk may be ameliorated by increasing fkbp5 mRNA expression in the amygdala during memory consolidation making this mechanism a plausible point of acute intervention to prevent the development of PTSD.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Transtornos de Estresse Pós-Traumáticos/genética , Transtornos de Estresse Pós-Traumáticos/metabolismo , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Adulto , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Nível de Alerta/efeitos dos fármacos , Nível de Alerta/fisiologia , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Feminino , Glucocorticoides/farmacologia , Humanos , Masculino , Rememoração Mental/efeitos dos fármacos , Rememoração Mental/fisiologia , Camundongos , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/metabolismo , Reflexo de Sobressalto/fisiologia , Transtornos de Estresse Pós-Traumáticos/psicologia
20.
Neuropsychopharmacology ; 41(11): 2714-22, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27238620

RESUMO

Recently we determined that activation of the tachykinin 2 (Tac2) pathway in the central amygdala (CeA) is necessary and sufficient for the modulation of fear memories. The Tac2 pathway includes the Tac2 gene, which encodes the neuropeptide neurokinin B and its corresponding receptor neurokinin 3 receptor (NK3R). In this study, using Tac2-cre and Tac2-GFP mice, we applied a combination of in vivo (optogenetics) and multiple in vitro techniques to further explore the mechanisms of action within the Tac2 pathway. In transgenic mice that express ChR2 solely in Tac2 neurons, in vivo optogenetic stimulation of CeA Tac2-expressing neurons during fear acquisition enhanced fear memory consolidation and drove action potential firing in vitro. In addition, Tac2-CeA neurons were shown to co-express striatal-enriched protein tyrosine phosphatase, which may have an important role in regulating Nk3R signaling during fear conditioning. These data extend our current understanding for the underlying mechanism(s) for the role of the Tac2 pathway in the regulation of fear memory, which may serve as a new therapeutic target in the treatment of fear-related disorders.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo , Aprendizagem/fisiologia , Precursores de Proteínas/genética , Transdução de Sinais/genética , Taquicininas/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Antipsicóticos/farmacologia , Channelrhodopsins , Condicionamento Clássico/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Locomoção/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organofosfatos/metabolismo , Piperidinas/farmacologia , Polímeros/metabolismo , Proteína Quinase C-delta/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Receptores da Neurocinina-3/genética , Receptores da Neurocinina-3/metabolismo , Taquicininas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...