Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38952926

RESUMEN

Background: Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. Methods: Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. Results: OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. Conclusions: SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.


Asunto(s)
Modelos Animales de Enfermedad , Extinción Psicológica , Miedo , Ratones Noqueados , Neuronas , Oxitocina , Síndrome de Prader-Willi , Somatostatina , Vasopresinas , Animales , Oxitocina/farmacología , Somatostatina/farmacología , Somatostatina/metabolismo , Miedo/efectos de los fármacos , Miedo/fisiología , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratones , Síndrome de Prader-Willi/fisiopatología , Síndrome de Prader-Willi/tratamiento farmacológico , Vasopresinas/metabolismo , Agresión/efectos de los fármacos , Agresión/fisiología , Masculino , Conducta Social , Núcleos Septales/efectos de los fármacos , Núcleos Septales/metabolismo , Optogenética , Ratones Endogámicos C57BL , Péptidos y Proteínas de Señalización Intracelular , Proteínas Intrínsecamente Desordenadas
2.
Philos Trans R Soc Lond B Biol Sci ; 377(1858): 20210057, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-35858094

RESUMEN

Oxytocin modulates social behaviour across diverse vertebrate taxa, but the precise nature of its effects varies across species, individuals and lifetimes. Contributing to this variation is the fact that oxytocin's physiological effects are mediated through interaction with diverse neuromodulatory systems and can depend on the specifics of the local circuits it acts on. Furthermore, those effects can be influenced by both genetics and experience. Here we discuss this complexity through the lens of a specific neuromodulatory system, endocannabinoids, interacting with oxytocin in the nucleus accumbens to modulate prosocial behaviours in prairie voles. We provide a survey of current knowledge of oxytocin-endocannabinoid interactions in relation to social behaviour. We review in detail recent research in monogamous female prairie voles demonstrating that social experience, such as mating and pair bonding, can change how oxytocin modulates nucleus accumbens glutamatergic signalling through the recruitment of endocannabinoids to modulate prosocial behaviour toward the partner. We then discuss potential sex differences in experience-dependent modulation of the nucleus accumbens by oxytocin in voles based on new data in males. Finally, we propose that future oxytocin-based precision medicine therapies should consider how prior social experience interacts with sex and genetics to influence oxytocin actions. This article is part of the theme issue 'Interplays between oxytocin and other neuromodulators in shaping complex social behaviours'.


Asunto(s)
Oxitocina , Apareamiento , Animales , Arvicolinae/metabolismo , Endocannabinoides , Femenino , Humanos , Masculino , Núcleo Accumbens/metabolismo , Receptores de Oxitocina/metabolismo , Conducta Social
3.
Curr Biol ; 32(5): 1026-1037.e4, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35108521

RESUMEN

Social relationships are dynamic and evolve with shared and personal experiences. Whether the functional role of social neuromodulators also evolves with experience to shape the trajectory of relationships is unknown. We utilized pair bonding in the socially monogamous prairie vole as an example of socio-sexual experience that dramatically alters behaviors displayed toward other individuals. We investigated oxytocin-dependent modulation of excitatory synaptic transmission in the nucleus accumbens as a function of pair-bonding status. We found that an oxytocin receptor agonist decreases the amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) in sexually naive virgin, but not pair-bonded, female voles, while it increases the amplitude of electrically evoked EPSCs in paired voles, but not in virgins. This oxytocin-induced potentiation of synaptic transmission relies on the de novo coupling between oxytocin receptor signaling and endocannabinoid receptor type 1 (CB1) receptor signaling in pair-bonded voles. Blocking CB1 receptors after pair-bond formation increases the occurrence of a specific form of social rejection-defensive upright response-that is displayed toward the partner, but not toward a novel individual. Altogether, our results demonstrate that oxytocin's action in the nucleus accumbens is changed through social experience in a way that regulates the trajectory of social interactions as the relationship with the partner unfolds, potentially promoting the maintenance of a pair bond by inhibiting aggressive responses. These results provide a mechanism by which social experience and context shift oxytocinergic signaling to impact neural and behavioral responses to social cues.


Asunto(s)
Núcleo Accumbens , Receptores de Oxitocina , Animales , Arvicolinae/metabolismo , Femenino , Pradera , Humanos , Núcleo Accumbens/metabolismo , Oxitocina/farmacología , Apareamiento , Receptores de Oxitocina/metabolismo , Conducta Social
5.
Handb Clin Neurol ; 182: 121-140, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34266588

RESUMEN

Oxytocin and vasopressin systems have been studied separately in autism spectrum disorder (ASD). Here, we provide evidence from an evolutionary and neuroscience perspective about the shared mechanisms and the common roles in regulating social behaviors. We first discuss findings on the evolutionary history of oxytocin and vasopressin ligands and receptors that highlight their common origin and clarify the evolutionary background of the crosstalk between them. Second, we conducted a comprehensive review of the increasing evidence for the role of both neuropeptides in regulating social behaviors. Third, we reviewed the growing evidence on the associations between the oxytocin/vasopressin systems and ASD, which includes oxytocin and vasopressin dysfunction in animal models of autism and in human patients, and the impact of treatments targeting the oxytocin or the vasopressin systems in children and in adults. Here, we highlight the potential of targeting the oxytocin/vasopressin systems to improve social deficits observed in ASD and the need for further investigations on how to transfer these research innovations into clinical applications.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Humanos , Oxitocina , Conducta Social , Vasopresinas
6.
Front Behav Neurosci ; 15: 814200, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35087387

RESUMEN

Impairments in social communication are common among neurodevelopmental disorders. While traditional animal models have advanced our understanding of the physiological and pathological development of social behavior, they do not recapitulate some aspects where social communication is essential, such as biparental care and the ability to form long-lasting social bonds. Prairie voles (Microtus ochrogaster) have emerged as a valuable rodent model in social neuroscience because they naturally display these behaviors. Nonetheless, the role of vocalizations in prairie vole social communication remains unclear. Here, we studied the ontogeny [from postnatal days (P) 8-16] of prairie vole pup ultrasonic vocalizations (USVs), both when isolated and when the mother was present but physically unattainable. In contrast to other similarly sized rodents such as mice, prairie vole pups of all ages produced isolation USVs with a relatively low fundamental frequency between 22 and 50 kHz, often with strong harmonic structure. Males consistently emitted vocalizations with a lower frequency than females. With age, pups vocalized less, and the acoustic features of vocalizations (e.g., duration and bandwidth) became more stereotyped. Manipulating an isolated pup's social environment by introducing its mother significantly increased vocal production at older (P12-16) but not younger ages, when pups were likely unable to hear or see her. Our data provide the first indication of a maturation in social context-dependent vocal emission, which may facilitate more active acoustic communication. These results help lay a foundation for the use of prairie voles as a model organism to probe the role of early life experience in the development of social-vocal communication.

7.
J Clin Invest ; 131(2)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33232306

RESUMEN

Intellectual and social disabilities are common comorbidities in adolescents and adults with MAGE family member L2 (MAGEL2) gene deficiency characterizing the Prader-Willi and Schaaf-Yang neurodevelopmental syndromes. The cellular and molecular mechanisms underlying the risk for autism in these syndromes are not understood. We asked whether vasopressin functions are altered by MAGEL2 deficiency and whether a treatment with vasopressin could alleviate the disabilities of social behavior. We used Magel2-knockout mice (adult males) combined with optogenetic or pharmacological tools to characterize disease modifications in the vasopressinergic brain system and monitor its impact on neurophysiological and behavioral functions. We found that the activation of vasopressin neurons and projections in the lateral septum were inappropriate for performing a social habituation/discrimination task. Mechanistically, the lack of vasopressin impeded the deactivation of somatostatin neurons in the lateral septum, which predicted social discrimination deficits. Correction of vasopressin septal content by administration or optogenetic stimulation of projecting axons suppressed the activity of somatostatin neurons and ameliorated social behavior. This preclinical study identified vasopressin in the lateral septum as a key factor in the pathophysiology of Magel2-related neurodevelopmental syndromes.


Asunto(s)
Antígenos de Neoplasias/genética , Trastorno Autístico , Conducta Animal , Proteínas/genética , Núcleos Septales , Conducta Social , Vasopresinas , Animales , Antígenos de Neoplasias/metabolismo , Trastorno Autístico/tratamiento farmacológico , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastorno Autístico/fisiopatología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Proteínas/metabolismo , Núcleos Septales/metabolismo , Núcleos Septales/fisiopatología , Vasopresinas/deficiencia , Vasopresinas/farmacología
8.
Proc Natl Acad Sci U S A ; 116(26): 13097-13106, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31182610

RESUMEN

Stress can either promote or impair learning and memory. Such opposing effects depend on whether synapses persist or decay after learning. Maintenance of new synapses formed at the time of learning upon neuronal network activation depends on the stress hormone-activated glucocorticoid receptor (GR) and neurotrophic factor release. Whether and how concurrent GR and neurotrophin signaling integrate to modulate synaptic plasticity and learning is not fully understood. Here, we show that deletion of the neurotrophin brain-derived neurotrophic factor (BDNF)-dependent GR-phosphorylation (PO4) sites impairs long-term memory retention and maintenance of newly formed postsynaptic dendritic spines in the mouse cortex after motor skills training. Chronic stress and the BDNF polymorphism Val66Met disrupt the BDNF-dependent GR-PO4 pathway necessary for preserving training-induced spines and previously acquired memories. Conversely, enrichment living promotes spine formation but fails to salvage training-related spines in mice lacking BDNF-dependent GR-PO4 sites, suggesting it is essential for spine consolidation and memory retention. Mechanistically, spine maturation and persistence in the motor cortex depend on synaptic mobilization of the glutamate receptor subunit A1 (GluA1) mediated by GR-PO4 Together, these findings indicate that regulation of GR-PO4 via activity-dependent BDNF signaling is important for the formation and maintenance of learning-dependent synapses. They also define a signaling mechanism underlying these effects.


Asunto(s)
Consolidación de la Memoria/fisiología , Corteza Motora/fisiopatología , Plasticidad Neuronal/fisiología , Receptores de Glucocorticoides/metabolismo , Estrés Psicológico/fisiopatología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ritmo Circadiano/fisiología , Espinas Dendríticas/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Glucocorticoides/metabolismo , Homeostasis/fisiología , Humanos , Microscopía Intravital , Masculino , Ratones , Corteza Motora/diagnóstico por imagen , Fosforilación/fisiología , Polimorfismo de Nucleótido Simple , Receptores AMPA/metabolismo , Receptores de Glucocorticoides/genética , Transducción de Señal/fisiología , Sinapsis/metabolismo
9.
Gen Comp Endocrinol ; 258: 15-32, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29155265

RESUMEN

It is now accepted that vasopressin, through V1A/V1B receptors, centrally regulates cognitive functions such as memory, affiliation, stress, fear and depression. However, the respective roles of these receptor isoforms and their contribution to stress-related pathologies remain uncertain. The development of new therapeutic treatments requires a precise knowledge of the distribution of these receptors within the brain, which has been so far hampered by the lack of selective V1B markers. In the present study, we have determined the pharmacological properties of three new potent rat V1B fluorescent ligands and demonstrated that they constitute valuable tools for simultaneous visualization and activation of native V1B receptors in living rat brain tissue. Thus, d[Leu4,Lys-Alexa 647)8]VP (analogue 3), the compound with the best affinity-selectivity/fluorescence ratio for the V1B receptor emerged as the most promising. The rat brain regions most concerned by stress such as hippocampus, olfactory bulbs, cortex and amygdala display the highest V1B fluorescent labelling with analogue 3. In the hippocampus CA2, V1B receptors are located on glutamatergic, not GABAergic neurones, and are absent from astrocytes. Using AVP-EGFP rats, we demonstrate the presence of V1B autoreceptors on AVP-secreting neurones not only in the hypothalamus, but also sparsely in the hippocampus. Finally, using both electrophysiology and visualization of ERK phosphorylation, we show analogue 3-induced activation of the V1B receptor in situ. This will help to analyse expression and functionality of V1B receptors in the brain and contribute to further explore the AVPergic circuitry in normal and pathological conditions.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/metabolismo , Colorantes Fluorescentes/metabolismo , Receptores de Vasopresinas/metabolismo , Animales , Arginina Vasopresina/metabolismo , Astrocitos/metabolismo , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Hipotálamo/metabolismo , Ligandos , Masculino , Neuroanatomía , Neuronas/metabolismo , Hipófisis/citología , Ratas Sprague-Dawley , Receptores de GABA/metabolismo , Coloración y Etiquetado , Vasopresinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...