Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 180(24): 3146-3159, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37482931

RESUMEN

BACKGROUND AND PURPOSE: Endocannabinoid (eCB) signalling gates many aspects of the stress response, including the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis is controlled by corticotropin releasing hormone (CRH) producing neurons in the paraventricular nucleus of the hypothalamus (PVN). Disruption of eCB signalling increases drive to the HPA axis, but the mechanisms subserving this process are poorly understood. EXPERIMENTAL APPROACH: Using an array of cellular, endocrine and behavioural readouts associated with activation of CRH neurons in the PVN, we evaluated the contributions of tonic eCB signalling to the generation of a stress response. KEY RESULTS: The CB1 receptor antagonist/inverse agonist AM251, neutral antagonist NESS243 and NAPE PLD inhibitor LEI401 all uniformly increased Fos in the PVN, unmasked stress-linked behaviours, such as grooming, and increased circulating CORT, recapitulating the effects of stress. Similar effects were also seen after direct administration of AM251 into the PVN, while optogenetic inhibition of PVN CRH neurons ameliorated stress-like behavioural changes produced by disruption of eCB signalling. CONCLUSIONS AND IMPLICATIONS: These data indicate that under resting conditions, constitutive eCB signalling restricts activation of the HPA axis through local regulation of CRH neurons in the PVN.


Asunto(s)
Endocannabinoides , Sistema Hipotálamo-Hipofisario , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Endocannabinoides/farmacología , Agonismo Inverso de Drogas , Sistema Hipófiso-Suprarrenal/metabolismo , Hipotálamo/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Núcleo Hipotalámico Paraventricular , Corticosterona/farmacología
2.
Elife ; 122023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37039453

RESUMEN

Fatty acid amide hydrolase (FAAH) degrades the endocannabinoid anandamide. A polymorphism in FAAH (FAAH C385A) reduces FAAH expression, increases anandamide levels, and increases the risk of obesity. Nevertheless, some studies have found no association between FAAH C385A and obesity. We investigated whether the environmental context governs the impact of FAAH C385A on metabolic outcomes. Using a C385A knock-in mouse model, we found that FAAH A/A mice are more susceptible to glucocorticoid-induced hyperphagia, weight gain, and activation of hypothalamic AMP-activated protein kinase (AMPK). AMPK inhibition occluded the amplified hyperphagic response to glucocorticoids in FAAH A/A mice. FAAH knockdown exclusively in agouti-related protein (AgRP) neurons mimicked the exaggerated feeding response of FAAH A/A mice to glucocorticoids. FAAH A/A mice likewise presented exaggerated orexigenic responses to ghrelin, while FAAH knockdown in AgRP neurons blunted leptin anorectic responses. Together, the FAAH A/A genotype amplifies orexigenic responses and decreases anorexigenic responses, providing a putative mechanism explaining the diverging human findings.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Endocannabinoides , Ratones , Humanos , Animales , Proteína Relacionada con Agouti , Endocannabinoides/metabolismo , Amidohidrolasas/metabolismo , Obesidad
3.
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
4.
Neuropsychopharmacology ; 45(5): 745-752, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31775159

RESUMEN

The endocannabinoid and dopaminergic systems have independently been implicated in substance use disorder and obesity. We investigated a potential interaction between genetically inherited variation in fatty acid amide hydrolase (FAAH, C385A), which metabolizes the cannabis-like endocannabinoid anandamide, and dopaminergic system, measured by dopamine receptor levels and mRNA. Binding of the dopamine D3 preferring probe [C-11]-(+)-PHNO was measured with positron emission tomography (PET) in 79 human subjects genotyped for the FAAH C385A polymorphism (36/79 AC + AA). Autoradiography with [H-3]-(+)-PHNO and in situ hybridization with a D3-specific S-35 riboprobe were carried out in 30 knock-in mice with the FAAH C385A polymorphism (20/30 AC + AA). We found that the FAAH genetic variant C385A was associated with significantly higher (+)-PHNO binding in both humans and in knock-in mice, and this effect was restricted to D3 selective brain regions (limbic striatum, globus pallidus, and ventral pallidum (9-14%; p < 0.04) in humans and Islands of Calleja (28%; p = 0.036) in mice). In situ hybridization with a D3-specific S-35 riboprobe in FAAH knock-in C385A mice confirmed significantly increased D3 receptor mRNA across examined regions (7-44%; p < 0.02). The association of reduced FAAH function with higher dopamine D3 receptors in human and mouse brain provide a mechanistic link between two brain systems that have been implicated in addiction-risk. This may explain the greater vulnerability for addiction and obesity in individuals with C385A genetic variant and by extension, suggest that a D3 antagonism strategy in substance use disorders should consider FAAH C385A polymorphism.


Asunto(s)
Amidohidrolasas/metabolismo , Encéfalo/metabolismo , Receptores de Dopamina D3/genética , Receptores de Dopamina D3/metabolismo , Trastornos Relacionados con Sustancias/enzimología , Adulto , Anciano , Animales , Autorradiografía , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación Missense , Polimorfismo de Nucleótido Simple , Tomografía de Emisión de Positrones , ARN Mensajero/metabolismo , Trastornos Relacionados con Sustancias/genética , Adulto Joven
5.
Mol Psychiatry ; 25(5): 993-1005, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-30120421

RESUMEN

Post-traumatic stress disorder (PTSD) is a common, debilitating condition with limited treatment options. Extinction of fear memories through prolonged exposure therapy, the primary evidence-based behavioral treatment for PTSD, has only partial efficacy. In mice, pharmacological inhibition of fatty acid amide hydrolase (FAAH) produces elevated levels of anandamide (AEA) and promotes fear extinction, suggesting that FAAH inhibitors may aid fear extinction-based treatments. A human FAAH 385C->A substitution encodes an FAAH enzyme with reduced catabolic efficacy. Individuals homozygous for the FAAH 385A allele may therefore offer a genetic model to evaluate the impact of elevations in AEA signaling in humans, helping to inform whether FAAH inhibitors have the potential to facilitate fear extinction therapy for PTSD. To overcome the challenge posed by low frequency of the AA genotype (appr. 5%), we prospectively genotyped 423 individuals to examine the balanced groups of CC, AC, and AA individuals (n = 25/group). Consistent with its loss-of-function nature, the A allele was dose dependently associated with elevated basal AEA levels, facilitated fear extinction, and enhanced the extinction recall. Moreover, the A-allele homozygotes were protected against stress-induced decreases in AEA and negative emotional consequences of stress. In a humanized mouse model, AA homozygous mice were similarly protected against stress-induced decreases in AEA, both in the periphery, and also in the amygdala and prefrontal cortex, brain structures critically involved in fear extinction and regulation of stress responses. Collectively, these data suggest that AEA signaling can temper aspects of the stress response and that FAAH inhibition may aid the treatment for stress-related psychiatric disorders, such as PTSD.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Endocannabinoides/metabolismo , Miedo , Alcamidas Poliinsaturadas/metabolismo , Trastornos por Estrés Postraumático/metabolismo , Trastornos por Estrés Postraumático/psicología , Adolescente , Adulto , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Sustancias Protectoras/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/genética , Adulto Joven
6.
Mol Metab ; 29: 170-181, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31668388

RESUMEN

BACKGROUND: Obesity, Type 2 diabetes (T2D) as well as stress-related disorders are rising public health threats and major burdens for modern society. Chronic stress and depression are highly associated with symptoms of the metabolic syndrome, but the molecular link is still not fully understood. Furthermore, therapies tackling these biological disorders are still lacking. The identification of shared molecular targets underlying both pathophysiologies may lead to the development of new treatments. The FK506 binding protein 51 (FKBP51) has recently been identified as a promising therapeutic target for stress-related psychiatric disorders and obesity-related metabolic outcomes. SCOPE OF THE REVIEW: The aim of this review is to summarize current evidence of in vitro, preclinical, and human studies on the stress responsive protein FKBP51, focusing on its newly discovered role in metabolism. Also, we highlight the therapeutic potential of FKBP51 as a new treatment target for symptoms of the metabolic syndrome. MAJOR CONCLUSIONS: We conclude the review by emphasizing missing knowledge gaps that remain and future research opportunities needed to implement FKBP51 as a drug target for stress-related obesity or T2D.


Asunto(s)
Enfermedades Metabólicas/patología , Estrés Psicológico , Proteínas de Unión a Tacrolimus/metabolismo , Adipogénesis , Animales , Metabolismo Energético , Glucosa/metabolismo , Humanos , Enfermedades Metabólicas/metabolismo , Obesidad/metabolismo , Obesidad/patología , Proteínas de Unión a Tacrolimus/genética
7.
Front Behav Neurosci ; 12: 262, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30483074

RESUMEN

Despite a growing body of research over the last few decades, mental disorders, including anxiety disorders or depression, are still one of the most prevalent and hardest to treat health burdens worldwide. Since pharmacological treatment with a single drug is often rather ineffective, approaches such as co-medication with functionally diverse antidepressants (ADs) have been discussed and tried more recently. Besides classical ADs, there is a growing number of candidate targets identified as potential starting points for new treatment methods. One of these candidates, the FK506 binding protein 51 (FKBP51) is linked to a number of psychiatric disorders in humans. In this study, we used SAFit2-a newly developed modulator of FKBP51, which has shown promising results in rodent models for stress-related disorders delivered in a depot formulation. We combined SAFit2 with the commonly prescribed selective serotonin reuptake inhibitor (SSRI) escitalopram and performed basic behavioral characterization in a mouse model. Remarkably, co-application of SAFit2 lowered the efficacy of escitalopram in anxiety-related tests but improved stress coping behavior. Given the fact that mental diseases such as anxiety disorders or depression can be divided into different sub-categories, some of which more or less prone to stress, SAFit2 could indeed be a highly beneficial co-medication in very specific cases. This study could be a first, promising step towards the use of FKBP51 modulators as potent and specific enhancers of AD efficiency for subclasses of patients in the future.

8.
Proc Natl Acad Sci U S A ; 115(29): 7605-7610, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29967158

RESUMEN

Endocannabinoid signaling regulates feeding and metabolic processes and has been linked to obesity development. Several hormonal signals, such as glucocorticoids and ghrelin, regulate feeding and metabolism by engaging the endocannabinoid system. Similarly, studies have suggested that leptin interacts with the endocannabinoid system, yet the mechanism and functional relevance of this interaction remain elusive. Therefore, we explored the interaction between leptin and endocannabinoid signaling with a focus on fatty acid amide hydrolase (FAAH), the primary degradative enzyme for the endocannabinoid N-arachidonoylethanolamine (anandamide; AEA). Mice deficient in leptin exhibited elevated hypothalamic AEA levels and reductions in FAAH activity while leptin administration to WT mice reduced AEA content and increased FAAH activity. Following high fat diet exposure, mice developed resistance to the effects of leptin administration on hypothalamic AEA content and FAAH activity. At a functional level, pharmacological inhibition of FAAH was sufficient to prevent leptin-mediated effects on body weight and food intake. Using a novel knock-in mouse model recapitulating a common human polymorphism (FAAH C385A; rs324420), which reduces FAAH activity, we investigated whether human genetic variance in FAAH affects leptin sensitivity. While WT (CC) mice were sensitive to leptin-induced reductions in food intake and body weight gain, low-expressing FAAH (AA) mice were unresponsive. These data demonstrate that FAAH activity is required for leptin's hypophagic effects and, at a translational level, suggest that a genetic variant in the FAAH gene contributes to differences in leptin sensitivity in human populations.


Asunto(s)
Amidohidrolasas/metabolismo , Ácidos Araquidónicos/metabolismo , Ingestión de Alimentos , Endocannabinoides/metabolismo , Metabolismo Energético/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/farmacología , Alcamidas Poliinsaturadas/metabolismo , Amidohidrolasas/genética , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Técnicas de Sustitución del Gen , Leptina/deficiencia , Masculino , Ratones , Ratones Noqueados , Polimorfismo Genético
9.
Neuropsychopharmacology ; 43(2): 342-353, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28540928

RESUMEN

Epigenetic regulation in anxiety is suggested, but evidence from large studies is needed. We conducted an epigenome-wide association study (EWAS) on anxiety in a population-based cohort and validated our finding in a clinical cohort as well as a murine model. In the KORA cohort, participants (n=1522, age 32-72 years) were administered the Generalized Anxiety Disorder (GAD-7) instrument, whole blood DNA methylation was measured (Illumina 450K BeadChip), and circulating levels of hs-CRP and IL-18 were assessed in the association between anxiety and methylation. DNA methylation was measured using the same instrument in a study of patients with anxiety disorders recruited at the Max Planck Institute of Psychiatry (MPIP, 131 non-medicated cases and 169 controls). To expand our mechanistic understanding, these findings were reverse translated in a mouse model of acute social defeat stress. In the KORA study, participants were classified according to mild, moderate, or severe levels of anxiety (29.4%/6.0%/1.5%, respectively). Severe anxiety was associated with 48.5% increased methylation at a single CpG site (cg12701571) located in the promoter of the gene encoding Asb1 (ß-coefficient=0.56 standard error (SE)=0.10, p (Bonferroni)=0.005), a protein hypothetically involved in regulation of cytokine signaling. An interaction between IL-18 and severe anxiety with methylation of this CpG cite showed a tendency towards significance in the total population (p=0.083) and a significant interaction among women (p=0.014). Methylation of the same CpG was positively associated with Panic and Agoraphobia scale (PAS) scores (ß=0.005, SE=0.002, p=0.021, n=131) among cases in the MPIP study. In a murine model of acute social defeat stress, Asb1 gene expression was significantly upregulated in a tissue-specific manner (p=0.006), which correlated with upregulation of the neuroimmunomodulating cytokine interleukin 1 beta. Our findings suggest epigenetic regulation of the stress-responsive Asb1 gene in anxiety-related phenotypes. Further studies are necessary to elucidate the causal direction of this association and the potential role of Asb1-mediated immune dysregulation in anxiety disorders.


Asunto(s)
Trastornos de Ansiedad/sangre , Trastornos de Ansiedad/fisiopatología , Islas de CpG/genética , Metilación de ADN , Epigénesis Genética/genética , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Adulto , Anciano , Animales , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Humanos , Interleucina-18/sangre , Interleucina-1beta/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Factores Sexuales , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología , Investigación Biomédica Traslacional
10.
Nat Commun ; 8(1): 1725, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-29170369

RESUMEN

The co-chaperone FKBP5 is a stress-responsive protein-regulating stress reactivity, and its genetic variants are associated with T2D related traits and other stress-related disorders. Here we show that FKBP51 plays a role in energy and glucose homeostasis. Fkbp5 knockout (51KO) mice are protected from high-fat diet-induced weight gain, show improved glucose tolerance and increased insulin signaling in skeletal muscle. Chronic treatment with a novel FKBP51 antagonist, SAFit2, recapitulates the effects of FKBP51 deletion on both body weight regulation and glucose tolerance. Using shorter SAFit2 treatment, we show that glucose tolerance improvement precedes the reduction in body weight. Mechanistically, we identify a novel association between FKBP51 and AS160, a substrate of AKT2 that is involved in glucose uptake. FKBP51 antagonism increases the phosphorylation of AS160, increases glucose transporter 4 expression at the plasma membrane, and ultimately enhances glucose uptake in skeletal myotubes. We propose FKBP51 as a mediator between stress and T2D development, and potential target for therapeutic approaches.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Transporte Biológico Activo , Dieta Alta en Grasa , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Fibras Musculares Esqueléticas/metabolismo , Fosforilación , Transducción de Señal , Estrés Fisiológico , Proteínas de Unión a Tacrolimus/deficiencia , Proteínas de Unión a Tacrolimus/genética , Aumento de Peso
11.
Front Neuroendocrinol ; 47: 86-108, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28739508

RESUMEN

For decades, there has been speculation regarding the interaction of cannabinoids with glucocorticoid systems. Given the functional redundancy between many of the physiological effects of glucocorticoids and cannabinoids, it was originally speculated that the biological mechanisms of cannabinoids were mediated by direct interactions with glucocorticoid systems. With the discovery of the endocannabinoid system, additional research demonstrated that it was actually the opposite; glucocorticoids recruit endocannabinoid signaling, and that the engagement of endocannabinoid signaling mediated many of the neurobiological and physiological effects of glucocorticoids. With the development of advances in pharmacology and genetics, significant advances in this area have been made, and it is now clear that functional interactions between these systems are critical for a wide array of physiological processes. The current review acts a comprehensive summary of the contemporary state of knowledge regarding the biological interactions between glucocorticoids and endocannabinoids, and their potential role in health and disease.


Asunto(s)
Endocannabinoides/metabolismo , Glucocorticoides/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo
12.
Psychoneuroendocrinology ; 78: 213-221, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28219813

RESUMEN

Chronic stress is a major risk factor for depression. Interestingly, not all individuals develop psychopathology after chronic stress exposure. In contrast to the prevailing view that stress effects are cumulative and increase stress vulnerability throughout life, the match/mismatch hypothesis of psychiatric disorders. The match/mismatch hypothesis proposes that individuals who experience moderate levels of early life psychosocial stress can acquire resilience to renewed stress exposure later in life. Here, we have tested this hypothesis by comparing the developmental effects of 2 opposite early life conditions, when followed by 2 opposite adult environments. Male Balb/c mice were exposed to either adverse early life conditions (limited nesting and bedding material) or a supportive rearing environment (early handling). At adulthood, the animals of each group were either housed with an ovariectomized female (supportive environment) or underwent chronic social defeat stress (socially adverse environment) for 3 weeks. At the end of the adult manipulations, all of the animals were returned to standard housing conditions. Then, we compared the neuroendocrine, behavioral and molecular effects of the interaction between early and adult environment. Our study shows that early life adversity does not necessarily result in increased vulnerability to stress. Specific endophenotypes, like hypothalamic-pituitary-adrenal axis activity, anxiety-related behavior and glucocorticoid receptor expression levels in the hippocampus were not significantly altered when adversity is experienced during early life and in adulthood, and are mainly affected by either early life or adult life adversity alone. Overall our data support the notion that being raised in a stressful environment prepares the offspring to better cope with a challenging adult environment and emphasize the role of early life experiences in shaping adult responsiveness to stress.


Asunto(s)
Adaptación Psicológica/fisiología , Conducta Animal/fisiología , Resiliencia Psicológica , Aislamiento Social , Estrés Psicológico/fisiopatología , Animales , Endofenotipos , Femenino , Hipocampo/metabolismo , Hipocampo/fisiopatología , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Ratones , Ratones Endogámicos BALB C , Sistema Hipófiso-Suprarrenal/fisiopatología , Receptores de Glucocorticoides/metabolismo , Medio Social
14.
Biol Psychiatry ; 80(10): 743-753, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27318500

RESUMEN

BACKGROUND: The medial prefrontal cortex (mPFC) subserves complex cognition and is impaired by stress. Corticotropin-releasing factor (CRF), through CRF receptor 1 (CRFR1), constitutes a key element of the stress response. However, its contribution to the effects of stress in the mPFC remains unclear. METHODS: Mice were exposed to acute social defeat stress and subsequently to either the temporal order memory (n = 11-12) or reversal learning (n = 9-11) behavioral test. Changes in mPFC Crhr1 messenger RNA levels were measured in acutely stressed mice (n = 12). Crhr1loxP/loxP mice received either intra-mPFC adeno-associated virus-Cre or empty microinjections (n = 17-20) and then were submitted to acute stress and later to the behavioral tests. Co-immunoprecipitation was used to detect activation of the protein kinase A (PKA) signaling pathway in the mPFC of acutely stressed mice (n = 8) or intra-mPFC CRF injected mice (n = 7). Finally, mice received intra-mPFC CRF (n = 11) and/or Rp-isomer cyclic adenosine 3',5' monophosphorothioate (Rp-cAMPS) (n = 12) microinjections and underwent behavioral testing. RESULTS: We report acute stress-induced effects on mPFC-mediated cognition, identify CRF-CRFR1-containing microcircuits within the mPFC, and demonstrate stress-induced changes in Crhr1 messenger RNA expression. Importantly, intra-mPFC CRFR1 deletion abolishes acute stress-induced executive dysfunction, whereas intra-mPFC CRF mimics acute stress-induced mPFC dysfunction. Acute stress and intra-mPFC CRF activate the PKA signaling pathway in the mPFC, leading to cyclic AMP response element binding protein phosphorylation in intra-mPFC CRFR1-expressing neurons. Finally, PKA blockade reverses the intra-mPFC CRF-induced executive dysfunction. CONCLUSIONS: Taken together, these results unravel a molecular mechanism linking acute stress to executive dysfunction via CRFR1. This will aid in the development of novel therapeutic targets for stress-induced cognitive dysfunction.


Asunto(s)
Disfunción Cognitiva/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Función Ejecutiva/fisiología , Corteza Prefrontal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Aprendizaje Inverso/fisiología , Estrés Psicológico/metabolismo , Enfermedad Aguda , Animales , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/fisiopatología , ARN Mensajero/metabolismo , Estrés Psicológico/complicaciones
15.
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
16.
Psychoneuroendocrinology ; 60: 138-50, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26143538

RESUMEN

Maternal diet during pregnancy can impact maternal behavior as well as the intrauterine environment, playing a critical role in programming offspring's physiology. In a preliminary study, we found a strong association between high-fat diet (HFD) during pregnancy and increased cannibalistic episodes and dams' mortality during late pregnancy and parturition. Based upon these data, we hypothesized that HFD during pregnancy could negatively affect neuroendocrine and metabolic regulations occurring during the final stages of pregnancy, thereby disrupting maternal behavior. To test this hypothesis, female C57BL/6J mice were fed HFD or control diet for 11 weeks until three days before the expected delivery date. Basal corticosterone plasma levels and brain levels of c-Fos were measured both before and after delivery, in addition to leptin levels in the adipose tissue. Dam's emotional behavior and social anxiety, in addition to locomotor activity were assessed before parturition. Data show that HFD led to aberrant maternal behavior, dams being characterized by behaviors related to aggression toward an unfamiliar social stimulus in the social avoidance test, in addition to decreased locomotor activity. Neural activity in HFD dams was reduced in the olfactory bulbs, a crucial brain region for social and olfactory recognition hence essential for maternal behavior. Furthermore, HFD feeding resulted in increased circulating levels of maternal corticosterone and decreased levels of leptin. In addition, the activity of the protective 11ß-dehydrogenase-2 (11ß-HSD-2) barrier in the placenta was decreased together with 11ß-dehydrogenase-1 (11ß-HSD-1) gene expression. Overall, these data suggest that HFD acts as a stressful challenge during pregnancy, impairing the neuroendocrine system and the neural activity of brain regions involved in the processing of relevant olfactory stimuli, with negative consequences on maternal physiology and behavior.


Asunto(s)
Encéfalo/fisiopatología , Dieta Alta en Grasa/efectos adversos , Glucocorticoides/fisiología , Conducta Materna , Estrés Psicológico/inducido químicamente , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/biosíntesis , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/biosíntesis , Animales , Química Encefálica/efectos de los fármacos , Canibalismo/psicología , Corticosterona/sangre , Ingestión de Energía , Femenino , Feto/metabolismo , Leptina/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Embarazo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología
17.
Neuron ; 86(5): 1189-202, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26050039

RESUMEN

Depression risk is exacerbated by genetic factors and stress exposure; however, the biological mechanisms through which these factors interact to confer depression risk are poorly understood. One putative biological mechanism implicates variability in the ability of cortisol, released in response to stress, to trigger a cascade of adaptive genomic and non-genomic processes through glucocorticoid receptor (GR) activation. Here, we demonstrate that common genetic variants in long-range enhancer elements modulate the immediate transcriptional response to GR activation in human blood cells. These functional genetic variants increase risk for depression and co-heritable psychiatric disorders. Moreover, these risk variants are associated with inappropriate amygdala reactivity, a transdiagnostic psychiatric endophenotype and an important stress hormone response trigger. Network modeling and animal experiments suggest that these genetic differences in GR-induced transcriptional activation may mediate the risk for depression and other psychiatric disorders by altering a network of functionally related stress-sensitive genes in blood and brain.


Asunto(s)
Encéfalo/fisiología , Variación Genética/genética , Trastornos Mentales/diagnóstico , Trastornos Mentales/genética , Estrés Psicológico/genética , Transcriptoma/genética , Animales , Estudios de Cohortes , Predicción , Redes Reguladoras de Genes/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Estrés Psicológico/diagnóstico
18.
Nat Chem Biol ; 11(1): 33-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25436518

RESUMEN

The FK506-binding protein 51 (FKBP51, encoded by the FKBP5 gene) is an established risk factor for stress-related psychiatric disorders such as major depression. Drug discovery for FKBP51 has been hampered by the inability to pharmacologically differentiate against the structurally similar but functional opposing homolog FKBP52, and all known FKBP ligands are unselective. Here, we report the discovery of the potent and highly selective inhibitors of FKBP51, SAFit1 and SAFit2. This new class of ligands achieves selectivity for FKBP51 by an induced-fit mechanism that is much less favorable for FKBP52. By using these ligands, we demonstrate that selective inhibition of FKBP51 enhances neurite elongation in neuronal cultures and improves neuroendocrine feedback and stress-coping behavior in mice. Our findings provide the structural and functional basis for the development of mechanistically new antidepressants.


Asunto(s)
Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Adaptación Psicológica/efectos de los fármacos , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Sitios de Unión/efectos de los fármacos , Células Cultivadas , Descubrimiento de Drogas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Neuritas/efectos de los fármacos , Conformación Proteica , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/efectos de los fármacos
19.
PLoS Med ; 11(11): e1001755, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25386878

RESUMEN

BACKGROUND: FK506 binding protein 51 (FKBP51) is an Hsp90 co-chaperone and regulator of the glucocorticoid receptor, and consequently of stress physiology. Clinical studies suggest a genetic link between FKBP51 and antidepressant response in mood disorders; however, the underlying mechanisms remain elusive. The objective of this study was to elucidate the role of FKBP51 in the actions of antidepressants, with a particular focus on pathways of autophagy. METHODS AND FINDINGS: Established cell lines, primary neural cells, human blood cells of healthy individuals and patients with depression, and mice were treated with antidepressants. Mice were tested for several neuroendocrine and behavioral parameters. Protein interactions and autophagic pathway activity were mainly evaluated by co-immunoprecipitation and Western blots. We first show that the effects of acute antidepressant treatment on behavior are abolished in FKBP51 knockout (51KO) mice. Autophagic markers, such as the autophagy initiator Beclin1, were increased following acute antidepressant treatment in brains from wild-type, but not 51KO, animals. FKBP51 binds to Beclin1, changes decisive protein interactions and phosphorylation of Beclin1, and triggers autophagic pathways. Antidepressants and FKBP51 exhibited synergistic effects on these pathways. Using chronic social defeat as a depression-relevant stress model in combination with chronic paroxetine (PAR) treatment revealed that the stress response, as well as the effects of antidepressants on behavior and autophagic markers, depends on FKBP51. In human blood cells of healthy individuals, FKBP51 levels correlated with the potential of antidepressants to induce autophagic pathways. Importantly, the clinical antidepressant response of patients with depression (n = 51) could be predicted by the antidepressant response of autophagic markers in patient-derived peripheral blood lymphocytes cultivated and treated ex vivo (Beclin1/amitriptyline: r = 0.572, p = 0.003; Beclin1/PAR: r = 0.569, p = 0.004; Beclin1/fluoxetine: r = 0.454, p = 0.026; pAkt/amitriptyline: r =  -0.416, p = 0.006; pAkt/PAR: r =  -0.355, p = 0.021; LC3B-II/PAR: r = 0.453, p = 0.02), as well as by the lymphocytic expression levels of FKBP51 (r = 0.631, p<0.0001), pAkt (r =  -0.515, p = 0.003), and Beclin1 (r = 0.521, p = 0.002) at admission. Limitations of the study include the use of male mice only and the relatively low number of patients for protein analyses. CONCLUSIONS: To our knowledge, these findings provide the first evidence for the molecular mechanism of FKBP51 in priming autophagic pathways; this process is linked to the potency of at least some antidepressants. These newly discovered functions of FKBP51 also provide novel predictive markers for treatment outcome, consistent with physiological and potential clinical relevance. Please see later in the article for the Editors' Summary.


Asunto(s)
Antidepresivos/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Depresión/genética , Trastorno Depresivo/genética , Estrés Psicológico/genética , Proteínas de Unión a Tacrolimus/genética , Adulto , Amitriptilina/farmacología , Amitriptilina/uso terapéutico , Animales , Antidepresivos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Células Sanguíneas/metabolismo , Depresión/tratamiento farmacológico , Depresión/metabolismo , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Femenino , Humanos , Leucocitos Mononucleares/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Paroxetina/farmacología , Paroxetina/uso terapéutico , Ratas Sprague-Dawley , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Adulto Joven
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...