Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Trauma Stress ; 37(5): 731-738, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39205469

RESUMEN

At the 39th meeting of the International Society of Traumatic Stress Studies, four leading scientists and clinicians were invited to reflect on their careers, focusing on the biological mechanisms and markers of traumatic stress. Dr. Raul Andero has contributed to understanding how stress alters memory networks in the brain, influencing the development of novel treatments. Dr. Tanja Jovanovic has pioneered the measurement and mechanistic understanding of fear learning, bridging basic and clinical research. Dr. Murray B. Stein has scaled up clinical and lab observations to large populations, refining the field's understanding of traumatic stress. Dr. Arieh Y Shalev has shaped the definition of traumatic stress, pioneering the longitudinal investigation of stress and integrating advanced computational methods to identify individuals at risk. These panelists were asked to reflect on their initial problems, ambitions, concerns, and unexpected challenges, as well as the influence of their work, on new research trajectories. Their insights provide valuable lessons about the process and content of their work, and their pioneering efforts have significantly advanced our understanding of the biological mechanisms and markers of traumatic stress.


Asunto(s)
Biomarcadores , Humanos , Salud Mental , Trastornos por Estrés Postraumático/psicología
2.
J Neurosci ; 41(5): 901-910, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33472824

RESUMEN

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.


Asunto(s)
Ansiedad/metabolismo , Emociones/fisiología , Sistema Límbico/metabolismo , Neuropéptidos/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Emociones/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Humanos , Sistema Límbico/efectos de los fármacos , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Neuropéptidos/farmacología , Neuropéptidos/uso terapéutico , Receptores de Taquicininas/antagonistas & inhibidores , Receptores de Taquicininas/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/psicología , Taquicininas/antagonistas & inhibidores , Taquicininas/metabolismo
3.
Mol Psychiatry ; 23(11): 2122-2132, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29298989

RESUMEN

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.


Asunto(s)
Miedo/fisiología , Memoria/fisiología , Investigación Biomédica Traslacional/tendencias , Animales , Ansiedad/terapia , Trastornos de Ansiedad/psicología , Extinción Psicológica/fisiología , Miedo/psicología , Humanos , Modelos Animales , Estrés Psicológico/fisiopatología , Investigación Biomédica Traslacional/métodos
4.
Cereb Cortex ; 28(4): 1233-1244, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28203747

RESUMEN

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.


Asunto(s)
Encéfalo/citología , Regulación de la Expresión Génica/fisiología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Fisiológico/fisiología , Hormona Adrenocorticotrópica/sangre , Animales , Autorradiografía , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
5.
Horm Behav ; 66(5): 713-23, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25311689

RESUMEN

In recent years, special attention is being paid to sex differences in susceptibility to disease. In this regard, there is evidence that male rats present higher levels of both cued and contextual fear conditioning than females. However, little is known about the concomitant hypothalamic-pituitary-adrenal (HPA) axis response to those situations which are critical in emotional memories. Here, we studied the behavioural and HPA responses of male and female Wistar rats to context fear conditioning using electric footshock as the aversive stimulus. Fear-conditioned rats showed a much greater ACTH and corticosterone response than those merely exposed to the fear conditioning chamber without receiving shocks. Moreover, males presented higher levels of freezing whereas HPA axis response was greater in females. Accordingly, during the fear extinction tests, female rats consistently showed less freezing and higher extinction rate, but greater HPA activation than males. Exposure to an open-field resulted in lower activity/exploration in fear-conditioned males, but not in females, suggesting greater conditioned cognitive generalization in males than females. It can be concluded that important sex differences in fear conditioning are observed in both freezing and HPA activation, but the two sets of variables are affected in the opposite direction: enhanced behavioural impact in males, but enhanced HPA responsiveness in females. Thus, the role of sex differences on fear-related stimuli may depend on the variables chosen to evaluate it, the greater responsiveness of the HPA axis in females perhaps being an important factor to be further explored.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Caracteres Sexuales , Animales , Corticosterona/metabolismo , Señales (Psicología) , Femenino , Masculino , Ratas , Ratas Wistar
6.
Psychoneuroendocrinology ; 170: 107164, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39146600

RESUMEN

Stress-related disorders are commonly associated with abnormalities in hypothalamic-pituitary-adrenal (HPA) axis activity. Preliminary studies with cortisol administration in the aftermath of trauma suggest that this HPA axis hormone can potentially prevent maladaptive behavioral and biological stress responses. However, the efficacy of glucocorticoid administration during the peripuberty period has not been tested yet, although this lifetime is a critical time window in brain development and is highly sensitive to the harmful effects of stress. To further examine the short and long-lasting impact of glucocorticoids treatment given during the post-peripubertal stress period, the present study utilized a rat model of peripubertal stress-induced psychopathology and animals were subjected to a battery of tests to assess anxiety-like behaviors, exploratory behavior and reactivity to novelty at late adolescence and sociability, anhedonia and stress coping behaviors at adulthood. All the experiments were performed in males and females to evaluate the potential behavioral sex differences. Overall, our results demonstrated that rats exposed to peripubertal stress show decreased sociability in adulthood without differences in anxiety and depression-like behaviors. Moreover, this study shows that the administration of corticosterone after stress exposure at peripuberty does not prevent stress-induced behavioral alterations. However, we observed that some stress-induced behavioural alterations and corticosterone responses are sex-specific. Thus, the data obtained highlight that delineating sex differences in stress-related studies may ultimately contribute to the development of effective therapeutic interventions for each sex.


Asunto(s)
Ansiedad , Conducta Animal , Corticosterona , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Estrés Psicológico , Animales , Corticosterona/sangre , Masculino , Ratas , Estrés Psicológico/metabolismo , Femenino , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Depresión/metabolismo , Depresión/prevención & control , Adaptación Psicológica/fisiología , Adaptación Psicológica/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Modelos Animales de Enfermedad , Anhedonia/fisiología , Anhedonia/efectos de los fármacos , Ratas Wistar
7.
Curr Neuropharmacol ; 22(5): 935-962, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37403395

RESUMEN

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.


Asunto(s)
Trastorno Depresivo Mayor , Trastornos por Estrés Postraumático , Animales , Humanos , Encéfalo/diagnóstico por imagen , Trastornos de Ansiedad , Biomarcadores , Imagen por Resonancia Magnética
8.
Sci Adv ; 10(28): eadk3365, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38985873

RESUMEN

It remains unexplored in the field of fear memory whether functional neuronal connectivity between two brain areas is necessary for one sex but not the other. Here, we show that chemogenetic silencing of centromedial (CeM)-Tac2 fibers in the lateral posterior BNST (BNSTpl) decreased fear memory consolidation in male mice but not females. Optogenetic excitation of CeM-Tac2 fibers in the BNSTpl exhibited enhanced inhibitory postsynaptic currents in males compared to females. In vivo calcium imaging analysis revealed a sex-dimorphic fear memory engram in the BNSTpl. Furthermore, in humans, the single-nucleotide polymorphism (SNP) in the Tac2 receptor (rs2765) (TAC3R) decreased CeM-BNST connectivity in a fear task, impaired fear memory consolidation, and increased the expression of the TAC3R mRNA in AA-carrier men but not in women. These sex differences in critical neuronal circuits underlying fear memory formation may be relevant to human neuropsychiatric disorders with fear memory alterations such as posttraumatic stress disorder.


Asunto(s)
Miedo , Memoria , Caracteres Sexuales , Miedo/fisiología , Animales , Femenino , Masculino , Humanos , Ratones , Memoria/fisiología , Polimorfismo de Nucleótido Simple , Adulto
9.
Neurosci Biobehav Rev ; 151: 105224, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37156310

RESUMEN

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.


Asunto(s)
Restricción Física , Roedores , Ratas , Ratones , Animales , Restricción Física/métodos , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal/fisiología , Corticosterona , Estrés Psicológico , Estrés Fisiológico
10.
Curr Top Behav Neurosci ; 64: 105-132, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37528309

RESUMEN

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.


Asunto(s)
Extinción Psicológica , Miedo , Femenino , Masculino , Humanos , Caracteres Sexuales , Hormonas Esteroides Gonadales/farmacología , Estradiol/farmacología
11.
Neuropharmacology ; 224: 109344, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36402246

RESUMEN

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'.


Asunto(s)
Glucocorticoides , Trastornos por Estrés Postraumático , Animales , Humanos , Glucocorticoides/uso terapéutico , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/prevención & control , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Ansiedad , Hidrocortisona
12.
bioRxiv ; 2023 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-37205515

RESUMEN

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.

13.
Hippocampus ; 22(3): 399-408, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21136519

RESUMEN

Post-traumatic stress disorder (PTSD) patients show cognitive deficits, but it is unclear whether these are a consequence of the pathology or a pre-existing factor of vulnerability to PTSD. Animal models may help to demonstrate whether or not exposure to certain stressors can actually induce long-lasting (LL; days) impairment of hippocampus-dependent memory tasks and to characterize neurobiological mechanisms. Adult male rats were exposed to 2-h immobilization on boards (IMO), a severe stressor, and spatial learning in the Morris water maze (MWM) was studied days later. Exposure to IMO did not modify learning or short-term memory in the MWM when learning started 3 or 9 days after IMO, but stressed rats did show impaired long-term memory at both times, in accordance with the severity of the stressor. New treatments to prevent PTSD symptoms are needed. Thus, considering the potential protective role of brain-derived neurotrophic factor (BDNF) on hippocampal function, 7,8-dihydroxyflavone (7,8-DHF), a recently characterized agonist of the BDNF receptor TrkB, was given before or after IMO in additional experiments. Again, exposure to IMO resulted in LL deficit in long-term memory, and such impairment was prevented by the administration of 7,8-DHF either 2 h prior IMO or 8 h after the termination of IMO. The finding that IMO-induced impairment of spatial memory was prevented by pharmacological potentiation of TrkB pathway with 7,8-DHF even when the drug was given 8 h after IMO suggests that IMO-induced impairment is likely to be a LL process that is strongly dependent on the integrity of the BDNF-TrkB system and is susceptible to poststress therapeutic interventions. 7,8-DHF may represent a new therapeutic approach for early treatment of subjects who have suffered traumatic experiences.


Asunto(s)
Flavanonas/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Memoria/efectos de los fármacos , Receptor trkB/agonistas , Estrés Fisiológico/fisiología , Estrés Psicológico/complicaciones , Hormona Adrenocorticotrópica/sangre , Animales , Corticosterona/sangre , Flavanonas/uso terapéutico , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Restricción Física , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/fisiopatología , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
14.
Biol Sex Differ ; 13(1): 28, 2022 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690790

RESUMEN

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.


Asunto(s)
Consolidación de la Memoria , Receptores Androgénicos , Receptores de Neuroquinina-3/metabolismo , Animales , Receptor beta de Estrógeno/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/farmacología , Masculino , Consolidación de la Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo
15.
Neurobiol Stress ; 18: 100448, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35685680

RESUMEN

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.

16.
Curr Protoc ; 1(5): e102, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33950571

RESUMEN

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.


Asunto(s)
Extinción Psicológica , Miedo , Animales , Femenino , Hormonas Esteroides Gonadales , Masculino , Ratones , Ratas , Roedores , Caracteres Sexuales
17.
Elife ; 102021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34787081

RESUMEN

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.


Asunto(s)
Memoria/fisiología , Biosíntesis de Proteínas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
18.
Genes Brain Behav ; 19(3): e12621, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31661603

RESUMEN

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.


Asunto(s)
Condicionamiento Clásico , Receptores de Neuropéptido/genética , Animales , Estimulación Eléctrica , Miedo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Neuropéptido/deficiencia , Factores Sexuales
19.
Nat Commun ; 11(1): 782, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32034128

RESUMEN

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.


Asunto(s)
Adicción a la Comida/fisiopatología , Núcleo Accumbens/fisiología , Corteza Prefrontal/fisiología , Receptores de Dopamina D2/genética , Animales , Modelos Animales de Enfermedad , Conducta Alimentaria/fisiología , Adicción a la Comida/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones Noqueados , Vías Nerviosas/fisiología , Receptor Cannabinoide CB1/genética , Transmisión Sináptica , Regulación hacia Arriba
20.
Neuropharmacology ; 144: 345-357, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30439419

RESUMEN

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.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Dronabinol/efectos adversos , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Psicotrópicos/efectos adversos , Estrés Psicológico/fisiopatología , Animales , Ansiedad/etiología , Ansiedad/patología , Ansiedad/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Espinas Dendríticas/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Miedo/psicología , Femenino , Masculino , Uso de la Marihuana/metabolismo , Uso de la Marihuana/patología , Uso de la Marihuana/psicología , Ratones Endogámicos C57BL , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Células Piramidales/fisiología , Maduración Sexual , Estrés Psicológico/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA