Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 173(1): 166-180.e14, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29502969

RESUMEN

Brain-wide fluctuations in local field potential oscillations reflect emergent network-level signals that mediate behavior. Cracking the code whereby these oscillations coordinate in time and space (spatiotemporal dynamics) to represent complex behaviors would provide fundamental insights into how the brain signals emotional pathology. Using machine learning, we discover a spatiotemporal dynamic network that predicts the emergence of major depressive disorder (MDD)-related behavioral dysfunction in mice subjected to chronic social defeat stress. Activity patterns in this network originate in prefrontal cortex and ventral striatum, relay through amygdala and ventral tegmental area, and converge in ventral hippocampus. This network is increased by acute threat, and it is also enhanced in three independent models of MDD vulnerability. Finally, we demonstrate that this vulnerability network is biologically distinct from the networks that encode dysfunction after stress. Thus, these findings reveal a convergent mechanism through which MDD vulnerability is mediated in the brain.


Asunto(s)
Encéfalo/fisiología , Depresión/patología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Depresión/fisiopatología , Modelos Animales de Enfermedad , Estimulación Eléctrica , Electrodos Implantados , Inmunoglobulina G/genética , Inmunoglobulina G/metabolismo , Ketamina/farmacología , Aprendizaje Automático , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenómenos Fisiológicos/efectos de los fármacos , Corteza Prefrontal/fisiología , Estrés Psicológico
2.
Proc Natl Acad Sci U S A ; 112(8): 2557-62, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25675490

RESUMEN

Brain serotonin (5-HT) deficiency and exposure to psychosocial stress have both been implicated in the etiology of depression and anxiety disorders, but whether 5-HT deficiency influences susceptibility to depression- and anxiety-like phenotypes induced by psychosocial stress has not been formally established. Most clinically effective antidepressants increase the extracellular levels of 5-HT, and thus it has been hypothesized that antidepressant responses result from the reversal of endogenous 5-HT deficiency, but this hypothesis remains highly controversial. Here we evaluated the impact of brain 5-HT deficiency on stress susceptibility and antidepressant-like responses using tryptophan hydroxylase 2 knockin (Tph2KI) mice, which display 60-80% reductions in brain 5-HT. Our results demonstrate that 5-HT deficiency leads to increased susceptibility to social defeat stress (SDS), a model of psychosocial stress, and prevents the fluoxetine (FLX)-induced reversal of SDS-induced social avoidance, suggesting that 5-HT deficiency may impair antidepressant responses. In light of recent clinical and preclinical studies highlighting the potential of inhibiting the lateral habenula (LHb) to achieve antidepressant and antidepressant-like responses, we also examined whether LHb inhibition could achieve antidepressant-like responses in FLX-insensitive Tph2KI mice subjected to SDS. Our data reveal that using designer receptors exclusively activated by designer drugs (DREADDs) to inhibit LHb activity leads to reduced SDS-induced social avoidance behavior in both WT and Tph2KI mice. This observation provides additional preclinical evidence that inhibiting the LHb might represent a promising alternative therapeutic approach under conditions in which selective 5-HT reuptake inhibitors are ineffective.


Asunto(s)
Antidepresivos/uso terapéutico , Encéfalo/metabolismo , Serotonina/deficiencia , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Animales , Antidepresivos/farmacología , Encéfalo/efectos de los fármacos , Susceptibilidad a Enfermedades , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Serotonina/metabolismo , Triptófano Hidroxilasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 109(15): 5838-43, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22460790

RESUMEN

Insulin resistance is a key factor in the etiology of type 2 diabetes. Insulin-stimulated glucose uptake is mediated by the glucose transporter 4 (GLUT4), which is expressed mainly in skeletal muscle and adipose tissue. Insulin-stimulated translocation of GLUT4 from its intracellular compartment to the plasma membrane is regulated by small guanosine triphosphate hydrolases (GTPases) and is essential for the maintenance of normal glucose homeostasis. Here we show that the p75 neurotrophin receptor (p75(NTR)) is a regulator of glucose uptake and insulin resistance. p75(NTR) knockout mice show increased insulin sensitivity on normal chow diet, independent of changes in body weight. Euglycemic-hyperinsulinemic clamp studies demonstrate that deletion of the p75(NTR) gene increases the insulin-stimulated glucose disposal rate and suppression of hepatic glucose production. Genetic depletion or shRNA knockdown of p75(NTR) in adipocytes or myoblasts increases insulin-stimulated glucose uptake and GLUT4 translocation. Conversely, overexpression of p75(NTR) in adipocytes decreases insulin-stimulated glucose transport. In adipocytes, p75(NTR) forms a complex with the Rab5 family GTPases Rab5 and Rab31 that regulate GLUT4 trafficking. Rab5 and Rab31 directly interact with p75(NTR) primarily via helix 4 of the p75(NTR) death domain. Adipocytes from p75(NTR) knockout mice show increased Rab5 and decreased Rab31 activities, and dominant negative Rab5 rescues the increase in glucose uptake seen in p75(NTR) knockout adipocytes. Our results identify p75(NTR) as a unique player in glucose metabolism and suggest that signaling from p75(NTR) to Rab5 family GTPases may represent a unique therapeutic target for insulin resistance and diabetes.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Resistencia a la Insulina , Receptor de Factor de Crecimiento Nervioso/metabolismo , Adipocitos/metabolismo , Secuencia de Aminoácidos , Animales , Peso Corporal , Transportador de Glucosa de Tipo 4/metabolismo , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Células Musculares/metabolismo , Músculo Esquelético/citología , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptor de Factor de Crecimiento Nervioso/química , Receptor de Factor de Crecimiento Nervioso/deficiencia , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
4.
J Neurosci ; 33(10): 4505-13, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23467366

RESUMEN

Although the majority of first-line antidepressants increase brain serotonin and rare polymorphisms in tryptophan hydroxlase-2 (Tph2), the rate-limiting enzyme in the brain serotonin synthesis pathway, have been identified in cohorts of subjects with major depressive disorder, the circuit level alterations that results from serotonergic hypofunction remain poorly understood. Here we use chronic multicircuit neurophysiological recordings to characterize functional interactions across cortical and limbic circuits in mice engineered to express a human loss-of-function depression allele Tph2-(R441H) [Tph2 knockin (Tph2KI)]. Our results show that Tph2KI mice exhibit increased intra-network synchrony within medial prefrontal cortex (mPFC) and basal amygdala (AMY) and increased inter-network synchrony between these two brain networks. Moreover, we demonstrate that chronic treatment with fluoxetine reverses several of the circuit alterations observed within Tph2KI mice. Together, our findings establish a functional link between functional hyposerotonergia and altered mPFC-AMY network dynamics.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Corteza Cerebral/fisiopatología , Depresión/genética , Depresión/patología , Vías Nerviosas/fisiopatología , Serotonina/deficiencia , Amígdala del Cerebelo/efectos de los fármacos , Análisis de Varianza , Animales , Arginina/genética , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/genética , Corteza Cerebral/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Modelos Animales de Enfermedad , Electrodos Implantados , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/genética , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Suspensión Trasera , Histidina/genética , Humanos , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/patología , Neuronas/fisiología , Serotonina/genética , Análisis Espectral , Triptófano Hidroxilasa/genética
5.
Int J Neuropsychopharmacol ; 18(4)2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25583694

RESUMEN

BACKGROUND: Chronic treatment with antidepressants has been shown to enhance neurogenesis in the adult mammalian brain. Although this effect was initially reported to be restricted to the hippocampus, recent work has suggested that fluoxetine, a selective serotonin reuptake inhibitor, also promotes neurogenesis in the cortex. However, whether antidepressants target neural progenitor cells in other brain regions has not been examined. METHODS: Here, we used BrdU labeling and immunohistochemistry with a transgenic mouse line in which nestin+ neural progenitor cells can be inducibly labeled with the fluorescent protein, Tomato, following tamoxifen administration. We investigated the effects of chronic fluoxetine on cell proliferation and nestin+ progenitor cells in periventricular areas in the medial hypothalamus and medial habenula, two brain areas involved in stress and anxiety responses. RESULTS: Our data provide the first in vivo evidence that fluoxetine promotes cell proliferation and neurogenesis and increases the mRNA levels of BDNF in the hypothalamus and habenula. CONCLUSIONS: By identifying novel cellular targets of fluoxetine, our results may provide new insight into the mechanisms underlying antidepressant responses.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Proliferación Celular/efectos de los fármacos , Fluoxetina/farmacología , Habénula/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Bromodesoxiuridina , Proliferación Celular/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Dominio Doblecortina , Habénula/citología , Habénula/fisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Hipotálamo/citología , Hipotálamo/fisiología , Inmunohistoquímica , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Nestina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuroglía/citología , Neuroglía/efectos de los fármacos , Neuroglía/fisiología , Neuropéptidos/metabolismo , ARN Mensajero/metabolismo
6.
Front Mol Neurosci ; 17: 1315366, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38486964

RESUMEN

Dopaminergic neurotransmission has emerged as a critical determinant of stress susceptibility and resilience. Although the dopamine transporter (DAT) is known to play a key role in maintaining dopamine (DA) homeostasis, its importance for the regulation of stress susceptibility remains largely unknown. Indeed, while numerous studies have examined the neurochemical and behavioral consequences of genetic loss of DAT, very few have compared responses to stress in wild-type and DAT-knockout (KO) animals. The current study compared the responses of male and female WT and DAT-KO mice to a model of sub-chronic stress. Our results reveal that DAT-KO mice are resistant to stress-induced increases in the latency to enter the light chamber of the light-dark emergence test and demonstrate that DAT-KO mice exhibit baseline reductions in forced swim test immobility and grooming time in the splash test of grooming behavior. In addition to these behavioral changes, our results highlight the importance of sex and dopaminergic neurotransmission on stress-induced changes in the expression and phosphorylation of several signal transduction molecules in the nucleus accumbens that have previously been implicated in the regulation of stress susceptibility, including ERK, GSK3ß, and ΔFosB. Overall, these results provide further evidence of the importance of dopaminergic neurotransmission in regulating stress susceptibility and suggest that genetic loss of DAT prevents stress-induced increases in anxiety-like behavior.

7.
Neuropharmacology ; 258: 110082, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39009217

RESUMEN

Exercise is known to reduce depression and anxiety symptoms. Although the cellular and molecular mechanisms underlying this effect remain unknown, exercise-induced increases in neurotransmitter release and hippocampal neurogenesis have been hypothesized to play key roles. One neurotransmitter that has been implicated in both antidepressant-like effects and the regulation of hippocampal neurogenesis is serotonin (5-HT). Complete loss of function of the brain 5-HT synthesis enzyme (tryptophan hydroxylase 2, Tph2) has been reported to prevent exercise-induced increases in neurogenesis and to block a subset of antidepressant-like responses to selective serotonin reuptake inhibitors (SSRIs), but whether partial loss of Tph2 function blocks the behavioral and neurogenic effects of exercise has not been established. This study used four tests that are predictive of antidepressant efficacy to determine the impact of 5-HT deficiency on responses to exercise in male and female mice. Our results demonstrate that low 5-HT impairs the behavioral effects of exercise in females in the forced swim and novelty-suppressed feeding tests. However, genetic reductions in 5-HT synthesis did not significantly impact exercise-induced alterations in cellular proliferation or immature neuron production in the hippocampus in either sex. These findings highlight the importance of brain 5-HT in mediating behavioral responses to exercise and suggest that individual differences in brain 5-HT synthesis could influence sensitivity to the mental health benefits of exercise. Furthermore, the observed disconnect between neurogenic and behavioral responses to exercise suggests that increased neurogenesis is unlikely to be the primary driver of the behavioral effects of exercise observed here.


Asunto(s)
Neurogénesis , Condicionamiento Físico Animal , Serotonina , Triptófano Hidroxilasa , Animales , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/genética , Neurogénesis/fisiología , Neurogénesis/efectos de los fármacos , Serotonina/metabolismo , Masculino , Femenino , Condicionamiento Físico Animal/fisiología , Ratones , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratones Transgénicos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Ratones Endogámicos C57BL , Técnicas de Sustitución del Gen , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología
8.
Int J Neuropsychopharmacol ; 16(9): 2081-94, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23672796

RESUMEN

Aberrant serotonin (5-HT) signalling and exposure to early life stress have both been suggested to play a role in anxiety- and impulsivity-related behaviours. However, whether congenital 5-HT deficiency × early life stress interactions influence the development of anxiety- or impulsivity-like behaviour has not been established. Here, we examined the effects of early life maternal separation (MS) stress on anxiety-like behaviour and behavioural disinhibition, a type of impulsivity-like behaviour, in wild-type (WT) and tryptophan hydroxylase 2 (Tph2) knock-in (Tph2KI) mice, which exhibit ~60-80% reductions in the levels of brain 5-HT due to a R439H mutation in Tph2. We also investigated the effects of 5-HT deficiency and early life stress on adult hippocampal neurogenesis, plasma corticosterone levels and several signal transduction pathways in the amygdala. We demonstrate that MS slightly increases anxiety-like behaviour in WT mice and induces behavioural disinhibition in Tph2KI animals. We also demonstrate that MS leads to a slight decrease in cell proliferation within the hippocampus and potentiates corticosterone responses to acute stress, but these effects are not affected by brain 5-HT deficiency. However, we show that 5-HT deficiency leads to significant alterations in SGK-1 and GSK3ß signalling and NMDA receptor expression in the amygdala in response to MS. Together, these findings support a potential role for 5-HT-dependent signalling in the amygdala in regulating the long-term effects of early life stress on anxiety-like behaviour and behavioural disinhibition.


Asunto(s)
Ansiedad/etiología , Conducta Animal , Encéfalo/metabolismo , Conducta Impulsiva/etiología , Serotonina/deficiencia , Estrés Psicológico/etiología , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/psicología , Ansiedad de Separación/complicaciones , Ansiedad de Separación/psicología , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Corticosterona/sangre , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Conducta Impulsiva/genética , Conducta Impulsiva/metabolismo , Conducta Impulsiva/fisiopatología , Conducta Impulsiva/psicología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora , Neurogénesis , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
9.
Physiol Behav ; 269: 114288, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37414236

RESUMEN

Pregnancy and the post-partum period are associated with substantial fluctuations in hormone levels and are frequently associated with significant stress. Many individuals also experience affective disturbances during the peri­partum period, including anxiety, the 'baby blues,' and post-partum depression. However, the extent to which these affective changes result from rapidly altering hormone levels, increased stress, or the combination of both remains largely unknown. The current study sought to evaluate the consequences of pregnancy-like hormonal changes on behavior and gene expression in c57BL/6 mice in the absence of stress using a hormone-simulated pregnancy model. Our results reveal that animals receiving hormone injections to simulate the high levels of estrogen observed in late pregnancy and animals withdrawn from estrogen to mimic the rapid decline in this hormone following parturition both exhibit increased anxiety-like behavior compared to ovariectomized controls in the novel open field test. However, no other significant anxiety- or depression-like alterations were observed in either hormone-treated group compared to ovariectomized controls. Both hormone administration and estrogen withdrawal were shown to induce several significant alterations in gene expression in the bed nucleus of the stria terminalis and the paraventricular nucleus of the hypothalamus. In contrast to the estrogen withdrawal hypothesis of post-partum depression, our results suggest that this method estrogen withdrawal following hormone-simulated pregnancy in the absence of stress does not induce phenotypes consistent with post-partum depression in c57BL/6 mice. However, given that estrogen withdrawal does lead to significant gene expression changes in two stress-sensitive brain regions, it remains possible that estrogen withdrawal could still contribute to affective dysregulation in the peri-partum period by influencing susceptibility to stress. Future research is required to evaluate this possibility.


Asunto(s)
Depresión Posparto , Humanos , Femenino , Ratones , Embarazo , Animales , Depresión Posparto/inducido químicamente , Depresión Posparto/genética , Depresión/inducido químicamente , Estrógenos/metabolismo , Periodo Posparto/psicología , Expresión Génica
10.
J Cell Biol ; 177(6): 1119-32, 2007 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-17576803

RESUMEN

Clearance of fibrin through proteolytic degradation is a critical step of matrix remodeling that contributes to tissue repair in a variety of pathological conditions, such as stroke, atherosclerosis, and pulmonary disease. However, the molecular mechanisms that regulate fibrin deposition are not known. Here, we report that the p75 neurotrophin receptor (p75(NTR)), a TNF receptor superfamily member up-regulated after tissue injury, blocks fibrinolysis by down-regulating the serine protease, tissue plasminogen activator (tPA), and up-regulating plasminogen activator inhibitor-1 (PAI-1). We have discovered a new mechanism in which phosphodiesterase PDE4A4/5 interacts with p75(NTR) to enhance cAMP degradation. The p75(NTR)-dependent down-regulation of cAMP results in a decrease in extracellular proteolytic activity. This mechanism is supported in vivo in p75(NTR)-deficient mice, which show increased proteolysis after sciatic nerve injury and lung fibrosis. Our results reveal a novel pathogenic mechanism by which p75(NTR) regulates degradation of cAMP and perpetuates scar formation after injury.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Fibrosis , Receptor de Factor de Crecimiento Nervioso/fisiología , Activador de Tejido Plasminógeno/antagonistas & inhibidores , Animales , Cicatriz/etiología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Fibrinólisis , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Inhibidor 1 de Activador Plasminogénico/genética , Nervio Ciático/lesiones , Heridas y Lesiones
11.
Front Behav Neurosci ; 16: 941884, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36172469

RESUMEN

Early life stress is known to increase the risk of depression and anxiety disorders, which are highly prevalent conditions that disproportionately affect women. However, the results of preclinical studies have been mixed, with some work suggesting that early life stress promotes anxiety-like behavior and/or increases susceptibility to subsequent stressors, and other research suggesting that early life stress reduces anxiety-like behavior and/or confers resilience to subsequent stress exposure. It is likely that factors such as sex and the timing and severity of early life and adult stress exposure dictate whether a particular early life experience promotes adaptive vs. maladaptive behavior later in life. Most work in this area has focused exclusively on males, but several sex differences in the effects of early life stress on subsequent stress susceptibility have been reported. The current study examined the impact of early life maternal separation on susceptibility to behavioral alterations induced by 3 days of variable stress in adulthood in male and female c57BL6 mice. Our results indicate that 3 days of adult stress is sufficient to increase anxiety-like behavior in several paradigms and to increase immobility in the forced swim test. In contrast, a history of maternal separation reduces anxiety-like behavior in several tests, particularly in males. These findings could contribute to our understanding of sex differences in mental illness by demonstrating that males are more likely than females to display adaptive responses to mild early life stressors.

12.
Behav Brain Res ; 425: 113811, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35219773

RESUMEN

Women are more likely than men to suffer from major depression and anxiety disorders, a fact that is thought to depend in part on sex differences in stress susceptibility. Consistent with this, several preclinical stress paradigms have been reported to exert differential effects in males vs. females. For example, several studies have reported that female rodents are susceptible to a subset of depression- and anxiety-like behaviors induced by six days of stress exposure while males remain largely resilient. The current study sought to evaluate the generalizability of this increased vulnerability of female mice to sub-chronic stressors by examining potential sex differences in response to a new five-day stress paradigm. In addition to measuring behavior, the current work also evaluated the effects of stress on the expression of several genes in the nucleus accumbens that have been suggested to underlie sex differences in behavioral responses to sub-chronic stress. The current results indicate that males and females exhibit mostly similar behavioral alterations after exposure to this new stress model, but several sex-specific molecular alterations were observed in the nucleus accumbens following stress. Overall, our data indicate that females do not exhibit a general increase in susceptibility to 'depression-' and 'anxiety-like' behaviors induced by sub-chronic stressors, and they could reflect an example of sexual convergence in which similar behavioral alterations occur in males and females despite sex-specific molecular changes.


Asunto(s)
Núcleo Accumbens , Estrés Psicológico , Animales , Ansiedad/metabolismo , Trastornos de Ansiedad/metabolismo , Femenino , Humanos , Masculino , Ratones , Núcleo Accumbens/metabolismo , Caracteres Sexuales , Estrés Psicológico/metabolismo
13.
Front Behav Neurosci ; 16: 958342, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36204485

RESUMEN

Stress is known to contribute to mental illness and alcohol use disorders, which are highly prevalent and lead to considerable disability. These stress-related disorders are characterized by significant sex differences, which remain poorly understood. Preclinical research comparing the effects of stress in males and females has the potential to provide new insights into the neurobiology of these conditions. The current study compared the effects of chronic and sub-chronic exposure to variable environmental stressors on binge-like alcohol consumption using the drinking-in-the-dark model in male and female c57BL6 mice. The results reveal that chronic, but not sub-chronic, exposure to variable stress increases alcohol intake in both sexes. Stress-induced alterations in gene expression were also compared in the nucleus accumbens, a brain region widely known to play a key role in stress susceptibility and reward processing. Real-time PCR data indicate that chronic, but not sub-chronic, environmental stress leads to downregulation of adenosine 2A (A2A) receptor mRNA. By contrast, sub-chronic stress increased CREB expression, while chronic stress did not. Several sex differences in the effects of stress on gene expression were also noted. Our results demonstrate that reductions in A2A receptor mRNA in the nucleus accumbens are associated with the increased binge drinking of chronically stressed animals, but future work will be required to determine the functional importance of this gene expression change. Continuing to define the molecular alterations associated with stress-induced increases in alcohol intake has the potential to provide insights into the development and progression of stress-related disorders.

14.
Psychopharmacology (Berl) ; 239(9): 2975-2984, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35750862

RESUMEN

RATIONALE: Although pharmacotherapies are often effective in reducing binge eating in conditions such as bulimia nervosa and binge eating disorder, subsets of patients do not benefit sufficiently from existing treatments, and the reasons for treatment failure remain unclear. OBJECTIVES: This study aimed to evaluate whether genetic reductions in brain serotonin influence binge eating and/or the ability of fluoxetine, a selective serotonin reuptake inhibitor, to reduce binge eating in mice. METHODS: This study used a validated model of binge-like consumption of high-fat diet to compare binge-like food intake in control and fluoxetine-treated wild-type and serotonin-deficient mice from the tryptophan hydroxylase 2 (R439H) knock-in line. In addition, real-time PCR was used to evaluate potential genotype and sex differences in the effects of fluoxetine on gene expression in the raphe nucleus. RESULTS: The results reveal that brain serotonin deficiency is sufficient to increase binge eating in males, but not females. However, while chronic fluoxetine reduced binge eating in both genotypes of males and in wild-type females, it failed to reduce binge eating in serotonin-deficient females. Transcriptional responses to chronic fluoxetine were also characterized by sex and genotype differences. CONCLUSIONS: Overall, this study revealed significant sex differences in the effects of fluoxetine and brain serotonin deficiency on binge-like food intake and suggests that low brain serotonin could impact eating disorders both by promoting binge eating and by limiting the efficacy of fluoxetine to reduce binge eating.


Asunto(s)
Trastorno por Atracón , Bulimia , Animales , Trastorno por Atracón/tratamiento farmacológico , Encéfalo/metabolismo , Bulimia/tratamiento farmacológico , Femenino , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Masculino , Ratones , Serotonina/metabolismo
15.
Front Neurosci ; 15: 683103, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276291

RESUMEN

Clinical studies have reported an increased risk of depression and anxiety disorders among individuals who are obese, and women are more likely than men to suffer from depression, anxiety, and obesity. However, the effects of obesity-promoting diets on depression- and anxiety-like behavior remain controversial. A recent study from our group used the tryptophan hydroxylase 2 (R439H) knock-in mouse line to evaluate the impact of genetic brain serotonin (5-HT) deficiency on behavioral responses to high fat diet (HFD) in male mice. That study indicated that chronic exposure to HFD induced pro-anxiety-like effects in the open field test and antidepressant-like effects in the forced swim test in wild-type males. Interestingly, the antidepressant-like effect of HFD, but not the anxiogenic effect, was blocked by brain 5-HT deficiency in males. The current work sought to repeat these studies in females. Our new data suggest that females are less susceptible than males to HFD-induced weight gain and HFD-induced alterations in behavior. In addition, the effects of chronic HFD on the expression of inflammation-related genes in the hippocampus were markedly different in females than we had previously reported in males, and HFD was shown to impact the expression of several inflammation-related genes in a genotype-dependent manner. Together, our findings highlight the importance of brain 5-HT and sex in regulating behavioral and molecular responses to HFD. Our results may have important implications for our understanding of the clinically observed sex differences in the consequences of obesity.

16.
J Sex Med ; 7(5): 1726-33, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20537061

RESUMEN

INTRODUCTION: The Manichean diagnosis, psychogenic or organic, is the first and most frequent diagnostic scope managing sexual disorders. The aim of this Controversy is to discuss if this philosophy is still useful both in the conceptual and clinical perspective. METHODS: Five scientists (an endocrinologist, two psychologist, and two urologists) with expertise in the area of psychosexology and sexual medicine were asked to contribute with their opinions. MAIN OUTCOME MEASURE: Expert opinion supported by the critical review of the currently available literature. RESULT: Expert # 1, who is Controversy's section Editor, suggests that the term psychogenic is redundant, because all sexual dysfunctions involve the mind and the relationship with (at least) one partner. Furthermore, he is strongly against the exclusion diagnosis, in agreement with the Expert # 5. The idea that the psychogenic etiology is always present is also sustained with good arguments by the Expert # 2. On the other hand, the two Experts # 3 and 4 argue that a pharmacological treatment such as type 5 phosphodiesterase inhibitors or dapoxetine work in both organic and psychogenic conditions and that the attempt to perform a diagnostic effort is frequently useless. Last but not least, the Expert # 5 concludes that the mind-body dualism is to be considered obsolete and unhelpful in a modern approach to the patient with a sexual dysfunction. CONCLUSIONS: The reader of the Journal will judge if there is still a room for the Manichean diagnosis of different sexual dysfunctions or if it is time to completely change our perspective on this essential aspect of clinical sexual medicine.


Asunto(s)
Disfunciones Sexuales Fisiológicas/diagnóstico , Disfunciones Sexuales Fisiológicas/etiología , Disfunciones Sexuales Psicológicas/diagnóstico , Disfunciones Sexuales Psicológicas/etiología , Terapia Cognitivo-Conductual , Terapia Combinada , Diagnóstico Diferencial , Disfunción Eréctil/diagnóstico , Disfunción Eréctil/etiología , Disfunción Eréctil/terapia , Femenino , Humanos , Masculino , Disfunciones Sexuales Fisiológicas/terapia , Disfunciones Sexuales Psicológicas/terapia , Resultado del Tratamiento
17.
Front Mol Neurosci ; 12: 298, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31920532

RESUMEN

Obesity is associated with an increased risk of depression and anxiety disorders, but the nature of the relationship(s) between obesity and mental illness remains highly controversial. Some argue that depression and anxiety lead to increased consumption of "comfort foods," the intake of which reduces negative affect and promotes obesity. In contrast, others have theorized that negative affect results from chronic excessive consumption of highly palatable foods. The brain serotonin (5-HT) system has long been implicated in both the development and treatment of mental illness. Preclinical studies have shown that low brain 5-HT exacerbates depression- and anxiety-like behaviors induced by stress and blocks reductions in depression-like behavior induced by antidepressants, but the effects of brain 5-HT deficiency on responses to high-fat diet (HFD) have not been explored. The current work used genetically modified mice to evaluate the effects of low 5-HT on behavioral and molecular alterations induced by chronic exposure to HFD. Our results reveal that HFD decreases depression-like behavior and increases some anxiety-like behaviors in wild-type (WT) mice. However, genetic brain 5-HT deficiency blocks HFD-induced reductions in forced swim immobility and prevents HFD-induced increases in hippocampal GSK3ß phosphorylation despite having no significant effects on HFD-induced changes in body weight or anxiety-like behavior. Together, our results suggest that brain 5-HT deficiency significantly impacts a subset of behavioral and molecular responses to HFD, a finding that could help explain the complex relationships between obesity and mental illness.

18.
Neuropsychopharmacology ; 44(12): 2082-2090, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31035282

RESUMEN

5-hydroxytryptophan (5-HTP) has shown therapeutic promise in a range of human CNS disorders. But native 5-HTP immediate release (IR) is poorly druggable, as rapid absorption causes rapid onset of adverse events, and rapid elimination causes fluctuating exposure. Recently, we reported that 5-HTP delivered as slow-release (SR) in mice augmented the brain pro-serotonergic effect of selective serotonin reuptake inhibitors (SSRIs), without the usual adverse events associated with 5-HTP IR. However, our previous study entailed translational limitations, in terms of route, dose, and duration. Here we modeled oral 5-HTP SR in mice by administering 5-HTP via the food. We modeled oral SSRI treatment via fluoxetine in the water, in a regimen recapitulating clinical pharmacokinetics and pharmacodynamics. 5-HTP SR produced plasma 5-HTP levels well within the range enhancing brain 5-HT function in humans. 5-HTP SR robustly increased brain 5-HT synthesis and levels. When administered with an SSRI, 5-HTP SR enhanced 5-HT-sensitive behaviors and neurotrophic mRNA expression. 5-HTP SR's pro-serotonergic effects were stronger in mice with endogenous brain 5-HT deficiency. In a comprehensive screen, 5-HTP SR was devoid of overt toxicological effects. The present preclinical data, appreciated in the context of published 5-HTP clinical data, suggest that 5-HTP SR could represent a new therapeutic approach to the plethora of CNS disorders potentially treatable with a pro-serotonergic drug. 5-HTP SR might in particular be therapeutically relevant when brain 5-HT deficiency is pathogenic and as an adjunctive augmentation therapy to SSRI therapy.


Asunto(s)
5-Hidroxitriptófano/farmacología , 5-Hidroxitriptófano/administración & dosificación , 5-Hidroxitriptófano/análisis , Administración Oral , Animales , Conducta Animal/efectos de los fármacos , Química Encefálica , Femenino , Fluoxetina/farmacología , Masculino , Ratones Transgénicos , Prueba de Estudio Conceptual , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
19.
Neuron ; 40(6): 1119-31, 2003 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-14687547

RESUMEN

The cerebellum provides an excellent system for understanding how afferent and target neurons coordinate sequential intercellular signals and cell-autonomous genetic programs in development. Mutations in the orphan nuclear receptor RORalpha block Purkinje cell differentiation with a secondary loss of afferent granule cells. We show that early transcriptional targets of RORalpha include both mitogenic signals for afferent progenitors and signal transduction genes required to process their subsequent synaptic input. RORalpha acts through recruitment of gene-specific sets of transcriptional cofactors, including beta-catenin, p300, and Tip60, but appears independent of CBP. One target promoter is Sonic hedgehog, and recombinant Sonic hedgehog restores granule precursor proliferation in RORalpha-deficient cerebellum. Our results suggest a link between RORalpha and beta-catenin pathways, confirm that a nuclear receptor employs distinct coactivator complexes at different target genes, and provide a logic for early RORalpha expression in coordinating expression of genes required for reciprocal signals in cerebellar development.


Asunto(s)
Señalización del Calcio/fisiología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Transactivadores/biosíntesis , Animales , Proteínas Hedgehog , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Células de Purkinje/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Transactivadores/genética
20.
Neuroscience ; 373: 122-136, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29341883

RESUMEN

The association between stress and mental illness has been well documented, but the molecular consequences of repeated exposure to stress have not been completely identified. The present study sought to elucidate the combinatorial effects of early-life maternal separation stress and adult social defeat stress on alterations in signal transduction and gene expression that have been previously implicated in susceptibility to psychosocial stress. Molecular analyses were performed in the prelimbic/infralimbic cortex, amygdala, and nucleus accumbens, three brain regions that have been suggested to play critical roles in determining stress responses. The current data reveal that both maternal separation and social defeat significantly impact the expression of genes involved in histone methylation and the ß-catenin-, endogenous opioid-, neurotrophin-, and glucocorticoid signaling pathways. Although the effects of maternal separation and social defeat were largely non-overlapping, a subset of genes in each brain region were governed by additive, opposing, or other types of interactions between these stress paradigms, thus highlighting potential molecular mechanisms through which these stressors might coordinately regulate brain function and behavior.


Asunto(s)
Encéfalo/metabolismo , Dominación-Subordinación , Privación Materna , Estrés Psicológico/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Masculino , Ratones Endogámicos C57BL , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA