Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(31): 12816-21, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858446

RESUMEN

The monoamine oxidase isoenzymes (MAOs) A and B play important roles in the homeostasis of monoaminergic neurotransmitters. The combined deficiency of MAO A and B results in significantly elevated levels of serotonin (5-hydroxytryptamine), norepinephrine, dopamine, and ß-phenylethylamine; in humans and mice, these neurochemical changes are accompanied by neurodevelopmental perturbations as well as autistic-like responses. Ample evidence indicates that normal levels of monoamines in the hippocampus, amygdala, frontal cortex, and cerebellum are required for the integrity of learning and memory. Thus, in the present study, the cognitive status of MAO A/B knockout (KO) mice was examined with a wide array of behavioral tests. In comparison with male wild-type littermates, MAO A/B KO mice exhibited abnormally high and overgeneralized fear conditioning and enhanced eye-blink conditioning. These alterations were accompanied by significant increases in hippocampal long-term potentiation and alterations in the relative expression of NMDA glutamate receptor subunits. Our data suggest that chronic elevations of monoamines, because of the absence of MAO A and MAO B, cause functional alterations that are accompanied with changes in the cellular mechanisms underlying learning and memory. The characteristics exhibited by MAO A/B KO mice highlight the potential of these animals as a useful tool to provide further insight into the molecular bases of disorders associated with abnormal monoaminergic profiles.


Asunto(s)
Conducta Animal , Monoaminas Biogénicas/metabolismo , Trastornos del Conocimiento , Hipocampo , Memoria , Monoaminooxidasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Animales , Trastornos del Conocimiento/enzimología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Hipocampo/enzimología , Hipocampo/patología , Hipocampo/fisiopatología , Humanos , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones Noqueados , Monoaminooxidasa/genética , Proteínas del Tejido Nervioso/genética , Neurotransmisores/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
2.
Int J Neuropsychopharmacol ; 18(9)2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25857821

RESUMEN

BACKGROUND: Acute stress triggers transient alterations in the synaptic release and metabolism of brain monoamine neurotransmitters. These rapid changes are essential to activate neuroplastic processes aimed at the appraisal of the stressor and enactment of commensurate defensive behaviors. Threat evaluation has been recently associated with the dendritic morphology of pyramidal cells in the orbitofrontal cortex (OFC) and basolateral amygdala (BLA); thus, we examined the rapid effects of restraint stress on anxiety-like behavior and dendritic morphology in the BLA and OFC of mice. Furthermore, we tested whether these processes may be affected by deficiency of monoamine oxidase A (MAO-A), the primary enzyme catalyzing monoamine metabolism. METHODS: Following a short-term (1-4h) restraint schedule, MAO-A knockout (KO) and wild-type (WT) mice were sacrificed, and histological analyses of dendrites in pyramidal neurons of the BLA and OFC of the animals were performed. Anxiety-like behaviors were examined in a separate cohort of animals subjected to the same experimental conditions. RESULTS: In WT mice, short-term restraint stress significantly enhanced anxiety-like responses, as well as a time-dependent proliferation of apical (but not basilar) dendrites of the OFC neurons; conversely, a retraction in BLA dendrites was observed. None of these behavioral and morphological changes were observed in MAO-A KO mice. CONCLUSIONS: These findings suggest that acute stress induces anxiety-like responses by affecting rapid dendritic remodeling in the pyramidal cells of OFC and BLA; furthermore, our data show that MAO-A and monoamine metabolism are required for these phenomena.


Asunto(s)
Ansiedad/enzimología , Complejo Nuclear Basolateral/patología , Dendritas/patología , Monoaminooxidasa/metabolismo , Corteza Prefrontal/patología , Células Piramidales/patología , Estrés Psicológico/enzimología , Animales , Ansiedad/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones de la Cepa 129 , Monoaminooxidasa/deficiencia , Estrés Psicológico/complicaciones
3.
Neuropsychopharmacology ; 49(9): 1373-1382, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38396257

RESUMEN

Persistence is the propensity to maintain goal-directed actions despite adversities. While this temperamental trait is crucial to mitigate depression risk, its neurobiological foundations remain elusive. Developing behavioral tasks to capture persistence in animal models is crucial for understanding its molecular underpinnings. Here, we introduce the Sinking Platform Test (SPT), a novel high-throughput paradigm to measure persistence. Mice were trained to exit a water-filled tank by ascending onto a platform above water level. Throughout the training, mice were also occasionally exposed to "failure trials," during which an operator would submerge a platform right after the mouse climbed onto it, requiring the mouse to reach and ascend a newly introduced platform. Following training, mice were subjected to a 5-min test exclusively consisting of failure trials. Male and female mice exhibited comparable persistence, measured by the number of climbed platforms during the test. Furthermore, this index was increased by chronic administration of fluoxetine or imipramine; conversely, it was reduced by acute and chronic haloperidol. Notably, six weeks of social isolation reduced SPT performance, and this effect was rescued by imipramine treatment over the last two weeks. A 4-week regimen of voluntary wheel running also improved persistence in socially isolated mice. Finally, comparing transcriptomic profiles of the prefrontal cortex of mice with high and low SPT performance revealed significant enrichment of immediate-early genes known to shape susceptibility for chronic stress. These findings highlight the potential of SPT as a promising method to uncover the biological mechanisms of persistence and evaluate novel interventions to enhance this response.


Asunto(s)
Fluoxetina , Haloperidol , Ratones Endogámicos C57BL , Aislamiento Social , Animales , Masculino , Ratones , Femenino , Fluoxetina/farmacología , Haloperidol/farmacología , Aislamiento Social/psicología , Imipramina/farmacología , Modelos Animales de Enfermedad , Conducta Animal/fisiología , Conducta Animal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología
4.
J Neurosci ; 32(25): 8574-82, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22723698

RESUMEN

Converging evidence shows that monoamine oxidase A (MAO A), the key enzyme catalyzing serotonin (5-hydroxytryptamine; 5-HT) and norepinephrine (NE) degradation, is a primary factor in the pathophysiology of antisocial and aggressive behavior. Accordingly, male MAO A-deficient humans and mice exhibit an extreme predisposition to aggressive outbursts in response to stress. As NMDARs regulate the emotional reactivity to social and environmental stimuli, we hypothesized their involvement in the modulation of aggression mediated by MAO A. In comparison with WT male mice, MAO A KO counterparts exhibited increases in 5-HT and NE levels across all brain regions, but no difference in glutamate concentrations and NMDAR binding. Notably, the prefrontal cortex (PFC) of MAO A KO mice exhibited higher expression of NR2A and NR2B, as well as lower levels of glycosylated NR1 subunits. In line with these changes, the current amplitude and decay time of NMDARs in PFC was significantly reduced. Furthermore, the currents of these receptors were hypersensitive to the action of the antagonists of the NMDAR complex (dizocilpine), as well as NR2A (PEAQX) and NR2B (Ro 25-6981) subunits. Notably, systemic administration of these agents selectively countered the enhanced aggression in MAO A KO mice, at doses that did not inherently affect motor activity. Our findings suggest that the role of MAO A in pathological aggression may be mediated by changes in NMDAR subunit composition in the PFC, and point to a critical function of this receptor in the molecular bases of antisocial personality.


Asunto(s)
Agresión/fisiología , Trastorno de Personalidad Antisocial/fisiopatología , Monoaminooxidasa/fisiología , Receptores de N-Metil-D-Aspartato/fisiología , Agresión/efectos de los fármacos , Animales , Autorradiografía , Sitios de Unión , Western Blotting , Cuerpo Estriado/metabolismo , Maleato de Dizocilpina/farmacología , Fenómenos Electrofisiológicos , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Hipocampo/metabolismo , Masculino , Ratones , Ratones Noqueados , Monoaminooxidasa/genética , Actividad Motora/fisiología , Norepinefrina/metabolismo , Técnicas de Placa-Clamp , Fenoles/farmacología , Piperidinas/farmacología , Prosencéfalo/enzimología , Quinoxalinas/farmacología , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Serotonina/metabolismo
5.
Int J Neuropsychopharmacol ; 16(5): 1059-70, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23174033

RESUMEN

Purinergic ionotropic P2X receptors are a family of cation-permeable channels that bind extracellular adenosine 5'-triphosphate. In particular, convergent lines of evidence have recently highlighted P2X(4) receptors as a potentially critical target in the regulation of multiple nervous and behavioural functions, including pain, neuroendocrine regulation and hippocampal plasticity. Nevertheless, the role of the P2X(4) receptor in behavioural organization remains poorly investigated. To study the effects of P2X(4) activation, we tested the acute effects of its potent positive allosteric modulator ivermectin (IVM, 2.5-10 mg/kg i.p.) on a broad set of paradigms capturing complementary aspects of perceptual, emotional and cognitive regulation in mice. In a novel open field, IVM did not induce significant changes in locomotor activity, but increased the time spent in the peripheral zone. In contrast, IVM produced anxiolytic-like effects in the elevated plus maze and marble burying tasks, as well as depression-like behaviours in the tail-suspension and forced swim tests. The agent induced no significant behavioural changes in the conditioned place preference test and in the novel object recognition task. Finally, the drug induced a dose-dependent decrease in sensorimotor gating, as assessed by pre-pulse inhibition (PPI) of the acoustic startle reflex. In P2X(4) knockout mice, the effects of IVM in the open field and elevated plus maze were similar to those observed in wild type mice; conversely, the drug significantly increased startle amplitude and failed to reduce PPI. Taken together, these results suggest that P2X(4) receptors may play a role in the regulation of sensorimotor gating.


Asunto(s)
Conducta Animal/efectos de los fármacos , Insecticidas/farmacología , Ivermectina/farmacología , Receptores Purinérgicos P2X4/metabolismo , Animales , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Suspensión Trasera , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dimensión del Dolor/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Receptores Purinérgicos P2X4/deficiencia , Filtrado Sensorial/efectos de los fármacos , Estadísticas no Paramétricas , Natación
6.
Int J Neuropsychopharmacol ; 16(4): 869-88, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22850464

RESUMEN

Converging lines of evidence show that a sizable subset of autism-spectrum disorders (ASDs) is characterized by increased blood levels of serotonin (5-hydroxytryptamine, 5-HT), yet the mechanistic link between these two phenomena remains unclear. The enzymatic degradation of brain 5-HT is mainly mediated by monoamine oxidase (MAO)A and, in the absence of this enzyme, by its cognate isoenzyme MAOB. MAOA and A/B knockout (KO) mice display high 5-HT levels, particularly during early developmental stages. Here we show that both mutant lines exhibit numerous behavioural hallmarks of ASDs, such as social and communication impairments, perseverative and stereotypical responses, behavioural inflexibility, as well as subtle tactile and motor deficits. Furthermore, both MAOA and A/B KO mice displayed neuropathological alterations reminiscent of typical ASD features, including reduced thickness of the corpus callosum, increased dendritic arborization of pyramidal neurons in the prefrontal cortex and disrupted microarchitecture of the cerebellum. The severity of repetitive responses and neuropathological aberrances was generally greater in MAOA/B KO animals. These findings suggest that the neurochemical imbalances induced by MAOA deficiency (either by itself or in conjunction with lack of MAOB) may result in an array of abnormalities similar to those observed in ASDs. Thus, MAOA and A/B KO mice may afford valuable models to help elucidate the neurobiological bases of these disorders and related neurodevelopmental problems.


Asunto(s)
Trastorno Autístico/enzimología , Relaciones Interpersonales , Aprendizaje por Laberinto/fisiología , Monoaminooxidasa/deficiencia , Actividad Motora/fisiología , Animales , Trastorno Autístico/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Monoaminooxidasa/genética , Actividad Motora/genética , Vocalización Animal/fisiología
7.
Sci Adv ; 8(47): eabm7069, 2022 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-36417527

RESUMEN

Little is understood about the embryonic development of sociality. We screened 1120 known drugs and found that embryonic inhibition of topoisomerase IIα (Top2a) resulted in lasting social deficits in zebrafish. In mice, prenatal Top2 inhibition caused defects in social interaction and communication, which are behaviors that relate to core symptoms of autism. Mutation of Top2a in zebrafish caused down-regulation of a set of genes highly enriched for genes associated with autism in humans. Both the Top2a-regulated and autism-associated gene sets have binding sites for polycomb repressive complex 2 (PRC2), a regulatory complex responsible for H3K27 trimethylation (H3K27me3). Moreover, both gene sets are highly enriched for H3K27me3. Inhibition of the PRC2 component Ezh2 rescued social deficits caused by Top2 inhibition. Therefore, Top2a is a key component of an evolutionarily conserved pathway that promotes the development of social behavior through PRC2 and H3K27me3.

8.
Int J Neuropsychopharmacol ; 14(9): 1195-207, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21156093

RESUMEN

Rich evidence indicates that monoamine oxidase (MAO) A, the major enzyme catalysing the degradation of monoamine neurotransmitters, plays a key role in emotional regulation. Although MAOA deficiency is associated with reactive aggression in humans and mice, the involvement of this enzyme in defensive behaviour remains controversial and poorly understood. To address this issue, we tested MAOA knockout (KO) mice in a spectrum of paradigms and settings associated with variable degrees of threat. The presentation of novel inanimate objects induced a significant reduction in exploratory approaches and increase in defensive behaviours, such as tail-rattling, biting and digging. These neophobic responses were context-dependent and particularly marked in the home cage. In the elevated plus- and T-mazes, MAOA KO mice and wild-type (WT) littermates displayed equivalent locomotor activity and time in closed and open arms; however, MAOA KO mice featured significant reductions in risk assessment, as well as unconditioned avoidance and escape. No differences between genotypes were observed in the defensive withdrawal and emergence test. Conversely, MAOA KO mice exhibited a dramatic reduction of defensive and fear-related behaviours in the presence of predator-related cues, such as predator urine or an anaesthetized rat, in comparison with those observed in their WT littermates. The behavioural abnormalities in MAOA KO mice were not paralleled by overt alterations in sensory and microvibrissal functions. Collectively, these results suggest that MAOA deficiency leads to a general inability to appropriately assess contextual risk and attune defensive and emotional responses to environmental cues.


Asunto(s)
Agresión , Conducta Animal , Monoaminooxidasa/deficiencia , Monoaminooxidasa/fisiología , Proteínas Mutantes/fisiología , Animales , Reacción de Prevención , Reacción de Fuga , Conducta Exploratoria , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/fisiología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Ratones Mutantes , Monoaminooxidasa/genética , Actividad Motora , Proteínas Mutantes/genética , Mutación Puntual , Distribución Aleatoria , Ratas , Ratas Long-Evans
9.
J Occup Environ Med ; 62(9): 746-756, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32890214

RESUMEN

OBJECTIVE: To describe caregiving work outcomes and related indirect (ie, productivity) and direct (ie, caregiving hours and expenses) costs. METHODS: A national, population-based survey to identify employed caregivers assisting a person with treatment-resistant depression (TRD) and a comparison group of employed caregivers assisting others (No TRD). RESULTS: Screening identified 169 TRD caregivers and 1070 No TRD caregivers providing 23.3 and 14.6 mean weekly caregiving hours, respectively. Adjusted annual indirect cost estimates were $11,121 for caregivers of TRD and $7761 for caregivers in the No TRD group (P ≤ 0.0001). At-work productivity loss (presenteeism) was the largest component. Adjusted annual direct cost estimates were $29,805 for caregivers of TRD and $20,642 for caregivers in the No TRD group (P ≤ 0.0001). CONCLUSIONS: TRD exacts a toll on caregivers and their employers exceeding that for other caregivers.


Asunto(s)
Cuidadores , Depresión/economía , Lugar de Trabajo , Costo de Enfermedad , Costos y Análisis de Costo , Eficiencia , Humanos , Presentismo
10.
Neuropharmacology ; 159: 107517, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30738037

RESUMEN

Aggressive behavior (AB) is a multifaceted disorder based on the interaction between genetic and environmental factors whose underlying mechanisms remain elusive. The best-characterized gene by environment (GxE) interaction for AB is the relationship between child neglect/abuse and low-activity alleles of the monoamine-oxidase A (MAOA) gene. MAOA oxidizes monoamines like serotonin and dopamine, whose aberrant signaling at discrete developmental ages plays a pivotal role in the ontogeny of AB. Here, we investigated the impact of this GxE on dopamine function at pre-adolescence by exposing hypomorphic MAOA (MAONeo) mice to early life stress (ES) and by performing behavioral and ex vivo electrophysiological analyses in the ventral tegmental area (VTA) and the prefrontal cortex (PFC). MAOANeo ES mouse dopamine neurons exhibited an enhanced post-synaptic responsiveness to excitatory inputs, aberrant plasticity in the PFC, and an AB. Systemic administration of the selective antagonist at dopamine D1 receptors SCH23390 fully restored PFC function and rescued AB. Collectively, these findings reveal that dysfunctional mesocortical dopamine signaling at pre-adolescence ties to AB in the MAOANeo ES mouse, and identify dopamine D1 receptor as a molecular target to be exploited for an age-tailored therapy. This article is part of the Special Issue entitled 'The neuropharmacology of social behavior: from bench to bedside'.


Asunto(s)
Agresión/fisiología , Neuronas Dopaminérgicas/metabolismo , Monoaminooxidasa/metabolismo , Red Nerviosa/metabolismo , Corteza Prefrontal/metabolismo , Estrés Psicológico/metabolismo , Área Tegmental Ventral/metabolismo , Factores de Edad , Agresión/efectos de los fármacos , Animales , Animales Recién Nacidos , Benzazepinas/farmacología , Dopamina/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Monoaminooxidasa/genética , Red Nerviosa/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Distribución Aleatoria , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/psicología , Área Tegmental Ventral/efectos de los fármacos
11.
Biomolecules ; 9(11)2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31752360

RESUMEN

Finasteride (FIN) is the prototypical inhibitor of steroid 5α-reductase (5αR), the enzyme that catalyzes the rate-limiting step of the conversion of progesterone and testosterone into their main neuroactive metabolites. FIN is clinically approved for the treatment of benign prostatic hyperplasia and male baldness; while often well-tolerated, FIN has also been shown to cause or exacerbate psychological problems in vulnerable subjects. Evidence on the psychological effects of FIN, however, remains controversial, in view of inconsistent clinical reports. Here, we tested the effects of FIN in a battery of tests aimed at capturing complementary aspects of mood regulation and stress reactivity in rats. FIN reduced exploratory, incentive, prosocial, and risk-taking behavior; furthermore, it decreased stress coping, as revealed by increased immobility in the forced-swim test (FST). This last effect was also observed in female and orchiectomized male rats, suggesting that the mechanism of action of FIN does not primarily reflect changes in gonadal steroids. The effects of FIN on FST responses were associated with a dramatic decrease in corticotropin release hormone (CRH) mRNA and adrenocorticotropic hormone (ACTH) levels. These results suggest that FIN impairs stress reactivity and reduces behavioral activation and impulsive behavior by altering the function of the hypothalamus-pituitary-adrenal (HPA) axis.


Asunto(s)
Inhibidores de 5-alfa-Reductasa/farmacología , Hormona Adrenocorticotrópica/metabolismo , Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/metabolismo , Finasterida/farmacología , Estrés Psicológico , Afecto/efectos de los fármacos , Animales , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Ratas , Ratas Long-Evans , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/patología
12.
Neuropharmacology ; 159: 107513, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30716416

RESUMEN

The ontogeny of antisocial behavior (ASB) is rooted in complex gene-environment (G×E) interactions. The best-characterized of these interplays occurs between: a) low-activity alleles of the gene encoding monoamine oxidase A (MAOA), the main serotonin-degrading enzyme; and b) child maltreatment. The purpose of this study was to develop the first animal model of this G×E interaction, to help understand the neurobiological mechanisms of ASB and identify novel targets for its therapy. Maoa hypomorphic transgenic mice were exposed to an early-life stress regimen consisting of maternal separation and daily intraperitoneal saline injections and were then compared with their wild-type and non-stressed controls for ASB-related neurobehavioral phenotypes. Maoa hypomorphic mice subjected to stress from postnatal day (PND) 1 through 7 - but not during the second postnatal week - developed overt aggression, social deficits and abnormal stress responses from the fourth week onwards. On PND 8, these mice exhibited low resting heart rate - a well-established premorbid sign of ASB - and a significant and selective up-regulation of serotonin 5-HT2A receptors in the prefrontal cortex. Notably, both aggression and neonatal bradycardia were rescued by the 5-HT2 receptor antagonist ketanserin (1-3 mg kg-1, IP), as well as the selective 5-HT2A receptor blocker MDL-100,907 (volinanserin, 0.1-0.3 mg kg-1, IP) throughout the first postnatal week. These findings provide the first evidence of a molecular basis of G×E interactions in ASB and point to early-life 5-HT2A receptor activation as a key mechanism for the ontogeny of this condition. This article is part of the Special Issue entitled 'The neuropharmacology of social behavior: from bench to bedside'.


Asunto(s)
Trastorno de Personalidad Antisocial/metabolismo , Interacción Gen-Ambiente , Privación Materna , Receptor de Serotonina 5-HT2A/metabolismo , Estrés Psicológico/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Trastorno de Personalidad Antisocial/psicología , Relación Dosis-Respuesta a Droga , Femenino , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Transgénicos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Estrés Psicológico/psicología
13.
Psychoneuroendocrinology ; 91: 95-104, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29544191

RESUMEN

The enzyme steroid 5α-reductase 2 (5αR2) catalyzes the conversion of testosterone into the potent androgen 5α-dihydrotestosterone. Previous investigations showed that 5αR2 is expressed in key brain areas for emotional and socio-affective reactivity, yet the role of this enzyme in behavioral regulation remains mostly unknown. Here, we profiled the behavioral characteristics of 5αR2 heterozygous (HZ) and knockout (KO) mice, as compared with their wild-type (WT) littermates. While male 5αR2 KO mice displayed no overt alterations in motoric, sensory, information-processing and anxiety-related behaviors, they exhibited deficits in neurobehavioral correlates of dominance (including aggression against intruders, mating, and tube dominance) as well as novelty-seeking and risk-taking responses. Furthermore, male 5αR2 KO mice exhibited reduced D2-like dopamine receptor binding in the shell of the nucleus accumbens - a well-recognized molecular signature of social dominance. Collectively, these results suggest that 5αR2 is involved in the establishment of social dominance and its behavioral manifestations. Further studies are warranted to understand how the metabolic actions of 5αR2 on steroid profile may be implicated in social ranking, impulse control, and the modulation of dopamine receptor expression in the nucleus accumbens.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/deficiencia , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/fisiología , Conducta Animal/fisiología , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Andrógenos/metabolismo , Animales , Dihidrotestosterona/metabolismo , Trastorno del Desarrollo Sexual 46,XY/psicología , Trastornos Disruptivos, del Control de Impulso y de la Conducta/fisiopatología , Conducta Exploratoria/efectos de los fármacos , Hipospadias/psicología , Masculino , Ratones , Ratones Noqueados , Predominio Social , Errores Congénitos del Metabolismo Esteroideo/psicología , Testosterona/metabolismo
14.
Neurosci Biobehav Rev ; 76(Pt A): 123-133, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27939782

RESUMEN

Tourette syndrome (TS) is a neurodevelopmental condition characterized by multiple, recurring motor and phonic tics. Rich empirical evidence shows that the severity of tics and associated manifestations is increased by several stressors and contextual triggers; however, the neurobiological mechanisms responsible for symptom exacerbation in TS remain poorly understood. This conceptual gap partially reflects the high phenotypic variability in tics, as well as the existing difficulties in operationalizing and standardizing stress and its effects in a clinical setting. Animal models of TS may be highly informative tools to overcome some of these limitations; these experimental preparations have already provided critical insights on key aspects of TS pathophysiology, and may prove useful to identify the neurochemical alterations induced by different stressful contingencies. In particular, emerging knowledge on the role of contextual triggers in animal models of TS may inform the development of novel pharmacological interventions to reduce tic fluctuations in this disorder.


Asunto(s)
Tics , Síndrome de Tourette , Animales , Humanos , Modelos Animales , Investigación Biomédica Traslacional
15.
J Neurosci Methods ; 292: 37-44, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28099872

RESUMEN

Tourette syndrome (TS) is a neurodevelopmental disorder characterized by multiple motor and phonic tics. While TS patients have been also shown to exhibit subtle abnormalities of sensorimotor integration and gait, animal models of this disorder are seldom tested for these functions. To fill this gap, we assessed gait and sensorimotor integration in the D1CT-7 mouse, one of the best-validated animal models of TS. D1CT-7 mice exhibit spontaneous tic-like manifestations, which, in line with the clinical phenomenology of TS, are markedly exacerbated by environmental stress. Thus, to verify whether stress may affect sensorimotor integration and gait functions in D1CT-7 mice, we subjected these animals to a 20-min session of spatial confinement, an environmental stressor that was recently shown to worsen tic-like manifestations. Immediately following this manipulation (or no confinement, for controls), animals were subjected to either the sticky-tape task, to test for sensorimotor integration; or a 60-min session in an open field (42×42cm) force-plate actometer for gait analysis. Gait analyses included spatial, temporal, and dynamic (force) parameters. D1CT-7 mice displayed a longer latency to remove a sticky tape, indicating marked impairments in sensorimotor integration; furthermore, these mutants exhibited shortened stride length, increased stride rate, nearly equal early-phase velocity, and higher late-phase velocity. D1CT-7 mice also ran with greater force amplitude than wild-type (WT) littermates. None of these phenotypes was worsened by spatial confinement. These results highlight the potential importance of testing sensorimotor integration and gait functions as a phenotypic correlate of cortical connectivity deficits in animal models of TS.


Asunto(s)
Trastornos Neurológicos de la Marcha/fisiopatología , Síndrome de Tourette/fisiopatología , Actigrafía , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Marcha , Masculino , Ratones Endogámicos BALB C , Ratones Mutantes , Destreza Motora
16.
Sci Rep ; 7(1): 3348, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28611376

RESUMEN

Tourette syndrome (TS) is a neuropsychiatric disorder characterized by multiple tics and sensorimotor abnormalities, the severity of which is typically increased by stress. The neurobiological underpinnings of this exacerbation, however, remain elusive. We recently reported that spatial confinement (SC), a moderate environmental stressor, increases tic-like responses and elicits TS-like sensorimotor gating deficits in the D1CT-7 mouse, one of the best-validated models of TS. Here, we hypothesized that these adverse effects may be mediated by neurosteroids, given their well-documented role in stress-response orchestration. Indeed, SC increased the levels of progesterone, as well as its derivatives 5α-dihydroprogesterone and allopregnanolone, in the prefrontal cortex (PFC) of D1CT-7 mice. Among these steroids, however, only allopregnanolone (5-15 mg/kg, IP) dose-dependently exacerbated TS-like manifestations in D1CT-7, but not wild-type littermates; these effects were countered by the benchmark anti-tic therapy haloperidol (0.3 mg/kg, IP). Furthermore, the phenotypic effects of spatial confinement in D1CT-7 mice were suppressed by finasteride (25-50 mg/kg, IP), an inhibitor of the main rate-limiting enzyme in allopregnanolone synthesis. These findings collectively suggest that stress may exacerbate TS symptoms by promoting allopregnanolone synthesis in the PFC, and corroborate previous clinical results pointing to finasteride as a novel therapeutic avenue to curb symptom fluctuations in TS.


Asunto(s)
Pregnanolona/metabolismo , Estrés Psicológico/metabolismo , Síndrome de Tourette/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Finasterida/farmacología , Haloperidol/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Pregnanolona/farmacología , Progesterona/metabolismo , Estrés Psicológico/fisiopatología , Síndrome de Tourette/fisiopatología
17.
Neuropharmacology ; 114: 77-87, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27889491

RESUMEN

Pramipexole (PPX) is a high-affinity D2-like dopamine receptor agonist, used in the treatment of Parkinson's disease (PD) and restless leg syndrome. Recent evidence indicates that PPX increases the risk of problem gambling and impulse-control disorders in vulnerable patients. Although the molecular bases of these complications remain unclear, several authors have theorized that PPX may increase risk propensity by activating presynaptic dopamine receptors in the mesolimbic system, resulting in the reduction of dopamine release in the nucleus accumbens (NAcc). To test this possibility, we subjected rats to a probability-discounting task specifically designed to capture the response to disadvantageous options. PPX enhanced disadvantageous decision-making at a dose (0.3 mg/kg/day, SC) that reduced phasic dopamine release in the NAcc. To test whether these modifications in dopamine efflux were responsible for the observed neuroeconomic deficits, PPX was administered in combination with the monoamine-depleting agent reserpine (RES), at a low dose (1 mg/kg/day, SC) that did not affect baseline locomotor and operant responses. Contrary to our predictions, RES surprisingly exacerbated the effects of PPX on disadvantageous decision-making, even though it failed to augment PPX-induced decreases in phasic dopamine release. These results collectively suggest that PPX impairs the discounting of probabilistic losses and that the enhancement in risk-taking behaviors secondary to this drug may be dissociated from dynamic changes in mesolimbic dopamine release.


Asunto(s)
Benzotiazoles/administración & dosificación , Toma de Decisiones/efectos de los fármacos , Toma de Decisiones/fisiología , Dopamina/fisiología , Núcleo Accumbens/fisiología , Receptores de Dopamina D2/agonistas , Asunción de Riesgos , Animales , Núcleo Caudado/metabolismo , Dopamina/metabolismo , Masculino , Norepinefrina/metabolismo , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Enfermedad de Parkinson/complicaciones , Pramipexol , Corteza Prefrontal/metabolismo , Probabilidad , Putamen/metabolismo , Ratas , Ratas Long-Evans , Serotonina/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-26776902

RESUMEN

Drawing upon the recent resurgence of biological criminology, several studies have highlighted a critical role for genetic factors in the ontogeny of antisocial and violent conduct. In particular, converging lines of evidence have documented that these maladaptive manifestations of aggression are influenced by monoamine oxidase A (MAOA), the enzyme that catalyzes the degradation of brain serotonin, norepinephrine and dopamine. The interest on the link between MAOA and aggression was originally sparked by Han Brunner's discovery of a syndrome characterized by marked antisocial behaviors in male carriers of a nonsense mutation of this gene. Subsequent studies showed that MAOA allelic variants associated with low enzyme activity moderate the impact of early-life maltreatment on aggression propensity. In spite of overwhelming evidence pointing to the relationship between MAOA and aggression, the neurobiological substrates of this link remain surprisingly elusive; very little is also known about the interventions that may reduce the severity of pathological aggression in genetically predisposed subjects. Animal models offer a unique experimental tool to investigate these issues; in particular, several lines of transgenic mice harboring total or partial loss-of-function Maoa mutations have been shown to recapitulate numerous psychological and neurofunctional endophenotypes observed in humans. This review summarizes the current knowledge on the link between MAOA and aggression; in particular, we will emphasize how an integrated translational strategy coordinating clinical and preclinical research may prove critical to elucidate important aspects of the pathophysiology of aggression, and identify potential targets for its diagnosis, prevention and treatment.


Asunto(s)
Agresión/fisiología , Monoaminooxidasa/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/patología , Humanos , Trastornos Mentales/enzimología , Trastornos Mentales/genética , Trastornos Mentales/patología , Monoaminooxidasa/genética
19.
Br J Pharmacol ; 173(13): 2111-21, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26171666

RESUMEN

BACKGROUND AND PURPOSE: The D1CT-7 mouse is one of the best known animal models of Tourette syndrome (TS), featuring spontaneous tic-like behaviours sensitive to standard TS therapies; these characteristics ensure a high face and predictive validity of this model, yet its construct validity remains elusive. To address this issue, we studied the responses of D1CT-7 mice to two critical components of TS pathophysiology: the exacerbation of tic-like behaviours in response to stress and the presence of sensorimotor gating deficits, which are thought to reflect the perceptual alterations causing the tics. EXPERIMENTAL APPROACH: D1CT-7 and wild-type (WT) littermates were subjected to a 20 min session of spatial confinement (SC) within an inescapable, 10 cm wide cylindrical enclosure. Changes in plasma corticosterone levels, tic-like behaviours and other spontaneous responses were measured. SC-exposed mice were also tested for the prepulse inhibition (PPI) of the startle response (a sensorimotor gating index) and other TS-related behaviours, including open-field locomotion, novel object exploration and social interaction and compared with non-confined counterparts. KEY RESULTS: SC produced a marked increase in corticosterone concentrations in both D1CT-7 and WT mice. In D1CT-7, but not WT mice, SC exacerbated tic-like and digging behaviours, and triggered PPI deficits and aggressive responses. Conversely, SC did not modify locomotor activity or novel object exploration in D1CT-7 mice. Both tic-like behaviours and PPI impairments in SC-exposed D1CT-7 mice were inhibited by standard TS therapies and D1 dopamine receptor antagonism. CONCLUSIONS AND IMPLICATIONS: These findings collectively support the translational and construct validity of D1CT-7 mice with respect to TS. LINKED ARTICLES: This article is part of a themed section on Updating Neuropathology and Neuropharmacology of Monoaminergic Systems. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.13/issuetoc.


Asunto(s)
Conducta Animal , Espacios Confinados , Modelos Animales de Enfermedad , Filtrado Sensorial , Síndrome de Tourette/fisiopatología , Animales , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
20.
Front Behav Neurosci ; 8: 71, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24639636

RESUMEN

Schizophrenia is a severe mental disorder, with a highly complex and heterogenous clinical presentation. Our current perspectives posit that the pathogenic mechanisms of this illness lie in complex arrays of gene × environment interactions. Furthermore, several findings indicate that males have a higher susceptibility for schizophrenia, with earlier age of onset and overall poorer clinical prognosis. Based on these premises, several authors have recently begun exploring the possibility that the greater schizophrenia vulnerability in males may reflect specific gene × sex (G×S) interactions. Our knowledge on such G×S interactions in schizophrenia is still rudimentary; nevertheless, the bulk of preclinical evidence suggests that the molecular mechanisms for such interactions are likely contributed by the neurobiological effects of sex steroids on dopamine (DA) neurotransmission. Accordingly, several recent studies suggest a gender-specific association of certain DAergic genes with schizophrenia. These G×S interactions have been particularly documented for catechol-O-methyltransferase (COMT) and monoamine oxidase (MAO), the main enzymes catalyzing DA metabolism. In the present review, we will outline the current evidence on the interactions of DA-related genes and sex-related factors, and discuss the potential molecular substrates that may mediate their cooperative actions in schizophrenia pathogenesis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA