Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cell Rep ; 42(8): 112874, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37516966

RESUMEN

Stress-related psychiatric disorders and the stress system show prominent differences between males and females, as well as strongly divergent transcriptional changes. Despite several proposed mechanisms, we still lack the understanding of the molecular processes at play. Here, we explore the contribution of cell types to transcriptional sex dimorphism using single-cell RNA sequencing. We identify cell-type-specific signatures of acute restraint stress in the paraventricular nucleus of the hypothalamus, a central hub of the stress response, in male and female mice. Further, we show that a history of chronic mild stress alters these signatures in a sex-specific way, and we identify oligodendrocytes as a major target for these sex-specific effects. This dataset, which we provide as an online interactive app, offers the transcriptomes of thousands of individual cells as a molecular resource for an in-depth dissection of the interplay between cell types and sex on the mechanisms of the stress response.


Asunto(s)
Caracteres Sexuales , Estrés Psicológico , Ratones , Masculino , Femenino , Animales , Estrés Psicológico/metabolismo , Hipotálamo
2.
Mol Metab ; 65: 101579, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36007872

RESUMEN

OBJECTIVE: Steroidogenic factor 1 (SF1) expressing neurons in the ventromedial hypothalamus (VMH) have been directly implicated in whole-body metabolism and in the onset of obesity. The co-chaperone FKBP51 is abundantly expressed in the VMH and was recently linked to type 2 diabetes, insulin resistance, adipogenesis, browning of white adipose tissue (WAT) and bodyweight regulation. METHODS: We investigated the role of FKBP51 in the VMH by conditional deletion and virus-mediated overexpression of FKBP51 in SF1-positive neurons. Baseline and high fat diet (HFD)-induced metabolic- and stress-related phenotypes in male and female mice were obtained. RESULTS: In contrast to previously reported robust phenotypes of FKBP51 manipulation in the entire mediobasal hypothalamus (MBH), selective deletion or overexpression of FKBP51 in the VMH resulted in only a moderate alteration of HFD-induced bodyweight gain and body composition, independent of sex. CONCLUSIONS: Overall, this study shows that animals lacking and overexpressing Fkbp5 in Sf1-expressing cells within the VMH display only a mild metabolic phenotype compared to an MBH-wide manipulation of this gene, suggesting that FKBP51 in SF1 neurons within this hypothalamic nucleus plays a subsidiary role in controlling whole-body metabolism.


Asunto(s)
Diabetes Mellitus Tipo 2 , Proteínas de Unión a Tacrolimus , Núcleo Hipotalámico Ventromedial , Animales , Diabetes Mellitus Tipo 2/metabolismo , Metabolismo Energético/fisiología , Femenino , Homeostasis/fisiología , Hipotálamo/metabolismo , Masculino , Ratones , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo
3.
Neuron ; 110(14): 2283-2298.e9, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35649415

RESUMEN

A single sub-anesthetic dose of ketamine produces a rapid and sustained antidepressant response, yet the molecular mechanisms responsible for this remain unclear. Here, we identified cell-type-specific transcriptional signatures associated with a sustained ketamine response in mice. Most interestingly, we identified the Kcnq2 gene as an important downstream regulator of ketamine action in glutamatergic neurons of the ventral hippocampus. We validated these findings through a series of complementary molecular, electrophysiological, cellular, pharmacological, behavioral, and functional experiments. We demonstrated that adjunctive treatment with retigabine, a KCNQ activator, augments ketamine's antidepressant-like effects in mice. Intriguingly, these effects are ketamine specific, as they do not modulate a response to classical antidepressants, such as escitalopram. These findings significantly advance our understanding of the mechanisms underlying the sustained antidepressant effects of ketamine, with important clinical implications.


Asunto(s)
Ketamina , Animales , Antidepresivos/farmacología , Hipocampo , Canal de Potasio KCNQ2/genética , Ketamina/farmacología , Ketamina/uso terapéutico , Ratones , Proteínas del Tejido Nervioso , Neuronas
4.
Sci Adv ; 8(10): eabi4797, 2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35263141

RESUMEN

The mediobasal hypothalamus (MBH) is the central region in the physiological response to metabolic stress. The FK506-binding protein 51 (FKBP51) is a major modulator of the stress response and has recently emerged as a scaffolder regulating metabolic and autophagy pathways. However, the detailed protein-protein interactions linking FKBP51 to autophagy upon metabolic challenges remain elusive. We performed mass spectrometry-based metabolomics of FKBP51 knockout (KO) cells revealing an increased amino acid and polyamine metabolism. We identified FKBP51 as a central nexus for the recruitment of the LKB1/AMPK complex to WIPI4 and TSC2 to WIPI3, thereby regulating the balance between autophagy and mTOR signaling in response to metabolic challenges. Furthermore, we demonstrated that MBH FKBP51 deletion strongly induces obesity, while its overexpression protects against high-fat diet (HFD)-induced obesity. Our study provides an important novel regulatory function of MBH FKBP51 within the stress-adapted autophagy response to metabolic challenges.


Asunto(s)
Hipotálamo , Proteínas de Unión a Tacrolimus , Autofagia , Dieta Alta en Grasa/efectos adversos , Humanos , Hipotálamo/metabolismo , Obesidad/metabolismo , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
5.
Mol Psychiatry ; 26(7): 3060-3076, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33649453

RESUMEN

Disturbed activation or regulation of the stress response through the hypothalamic-pituitary-adrenal (HPA) axis is a fundamental component of multiple stress-related diseases, including psychiatric, metabolic, and immune disorders. The FK506 binding protein 51 (FKBP5) is a negative regulator of the glucocorticoid receptor (GR), the main driver of HPA axis regulation, and FKBP5 polymorphisms have been repeatedly linked to stress-related disorders in humans. However, the specific role of Fkbp5 in the paraventricular nucleus of the hypothalamus (PVN) in shaping HPA axis (re)activity remains to be elucidated. We here demonstrate that the deletion of Fkbp5 in Sim1+ neurons dampens the acute stress response and increases GR sensitivity. In contrast, Fkbp5 overexpression in the PVN results in a chronic HPA axis over-activation, and a PVN-specific rescue of Fkbp5 expression in full Fkbp5 KO mice normalizes the HPA axis phenotype. Single-cell RNA sequencing revealed the cell-type-specific expression pattern of Fkbp5 in the PVN and showed that Fkbp5 expression is specifically upregulated in Crh+ neurons after stress. Finally, Crh-specific Fkbp5 overexpression alters Crh neuron activity, but only partially recapitulates the PVN-specific Fkbp5 overexpression phenotype. Together, the data establish the central and cell-type-specific importance of Fkbp5 in the PVN in shaping HPA axis regulation and the acute stress response.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Núcleo Hipotalámico Paraventricular , Estrés Fisiológico , Proteínas de Unión a Tacrolimus , Animales , Corticosterona , Hormona Liberadora de Corticotropina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proteínas de Unión a Tacrolimus/genética
6.
J Clin Invest ; 130(11): 6093-6108, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32780722

RESUMEN

Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Tipo 2/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Hipotálamo/enzimología , Resistencia a la Insulina , MAP Quinasa Quinasa 4/metabolismo , Transducción de Señal , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 2/genética , Fosfatasas de Especificidad Dual/genética , MAP Quinasa Quinasa 4/genética , Ratones , Ratones Noqueados
7.
Neuroendocrinology ; 103(3-4): 354-68, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26279463

RESUMEN

BACKGROUND: There is growing evidence that maternal obesity and prenatal exposure to a high-fat diet program fetal development to regulate the physiology and behavior of the offspring in adulthood. Yet the extent to which the maternal dietary environment contributes to adult disease vulnerability remains unclear. In the current study we tested whether prenatal exposure to maternal obesity increases the offspring's vulnerability to stress-related psychiatric disorders. METHODS: We used a mouse model of maternal diet-induced obesity to investigate whether maternal obesity affects the response to adult chronic stress exposure in young adult (3-month-old) and aged adult (12-month-old) offspring. RESULTS: Long-lasting, delayed impairments to anxiety-like behaviors and stress coping strategies resulted on account of prenatal exposure to maternal obesity. Although maternal obesity did not change the offspring's behavioral response to chronic stress per se, we demonstrate that the behavioral outcomes induced by prenatal exposure to maternal obesity parallel the deleterious effects of adult chronic stress exposure in aged male mice. We found that the glucocorticoid receptor (GR, Nr3c1) is upregulated in various hypothalamic nuclei on account of maternal obesity. In addition, gene expression of a known regulator of the GR, FKBP51, is increased specifically within the paraventricular nucleus. CONCLUSIONS: These findings indicate that maternal obesity parallels the deleterious effects of adult chronic stress exposure, and furthermore identifies GR/FKBP51 signaling as a novel candidate pathway regulated by maternal obesity.


Asunto(s)
Adaptación Psicológica/fisiología , Ansiedad/etiología , Dieta Alta en Grasa/efectos adversos , Obesidad/etiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Estrés Psicológico/etiología , Factores de Edad , Envejecimiento/psicología , Animales , Ansiedad/patología , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo , Receptores de Glucocorticoides/deficiencia , Receptores de Glucocorticoides/metabolismo , Natación/psicología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
8.
J Endocrinol ; 222(1): 15-26, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24781256

RESUMEN

While it is known that stress promotes obesity, the effects of stress within an obesogenic context are not so clear and molecular targets at the interface remain elusive. The FK506-binding protein 51 (FKBP51, gene: Fkbp5) has been identified as a target gene implicated in the development of stress-related psychiatric disorders and is a possible candidate for involvement in stress and metabolic regulation. The aims of the current study are to investigate the interaction between chronic stress and an obesogenic context and to additionally examine whether FKBP51 is involved in this interaction. For this purpose, male C57BL/6 mice were exposed to a high-fat diet for 8 weeks before being challenged with chronic social defeat stress. Herein, we demonstrate that chronic stress induces hypophagia and weight loss, ultimately improving features arising from an obesogenic context, including glucose tolerance and levels of insulin and leptin. We show that Fkbp5 expression is responsive to diet and stress in the hypothalamus and hippocampus respectively. Furthermore, under basal conditions, higher levels of hypothalamic Fkbp5 expression were related to increased body weight gain. Our data indicate that Fkbp5 may represent a novel target in metabolic regulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Obesidad/etiología , Obesidad/fisiopatología , Estrés Psicológico/fisiopatología , Proteínas de Unión a Tacrolimus/fisiología , Animales , Corticosterona/sangre , Modelos Animales de Enfermedad , Metabolismo Energético/fisiología , Glucosa/metabolismo , Hipocampo/fisiología , Hipotálamo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Aumento de Peso/fisiología
9.
J Neurosci ; 30(27): 9103-16, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20610744

RESUMEN

Urocortin 3 (UCN3) is strongly expressed in specific nuclei of the rodent brain, at sites distinct from those expressing urocortin 1 and urocortin 2, the other endogenous ligands of corticotropin-releasing hormone receptor type 2 (CRH-R2). To determine the physiological role of UCN3, we generated UCN3-deficient mice, in which the UCN3 open reading frame was replaced by a tau-lacZ reporter gene. By means of this reporter gene, the nucleus parabrachialis and the premammillary nucleus were identified as previously unknown sites of UCN3 expression. Additionally, the introduced reporter gene enabled the visualization of axonal projections of UCN3-expressing neurons from the superior paraolivary nucleus to the inferior colliculus and from the posterodorsal part of the medial amygdala to the principal nucleus of the bed nucleus of the stria terminalis, respectively. The examination of tau-lacZ reporter gene activity throughout the brain underscored a predominant expression of UCN3 in nuclei functionally connected to the accessory olfactory system. Male and female mice were comprehensively phenotyped but none of the applied tests provided indications for a role of UCN3 in the context of hypothalamic-pituitary-adrenocortical axis regulation, anxiety- or depression-related behavior. However, inspired by the prevalent expression throughout the accessory olfactory system, we identified alterations in social discrimination abilities of male and female UCN3 knock-out mice that were also present in male CRH-R2 knock-out mice. In conclusion, our results suggest a novel role for UCN3 and CRH-R2 related to the processing of social cues and to the establishment of social memories.


Asunto(s)
Discriminación en Psicología/fisiología , Relaciones Interpersonales , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Reconocimiento en Psicología/fisiología , Urocortinas/metabolismo , Estimulación Acústica/métodos , Animales , Encéfalo/citología , Encéfalo/metabolismo , Ritmo Circadiano/fisiología , Corticosterona/sangre , Miedo/fisiología , Femenino , Regulación de la Expresión Génica/genética , Sistema Hipotálamo-Hipofisario/metabolismo , Inhibición Psicológica , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Odorantes , Vías Olfatorias/fisiología , Percepción/fisiología , Sistema Hipófiso-Suprarrenal/embriología , Radioinmunoensayo/métodos , Receptores de Hormona Liberadora de Corticotropina/deficiencia , Reflejo de Sobresalto/genética , Estadísticas no Paramétricas , Natación/fisiología , Urocortinas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA