Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923479

RESUMEN

Schizophrenia is a complex psychopathology whose treatment is still challenging. Given the limitations of existing antipsychotics, there is urgent need for novel drugs with fewer side effects. SEP-363856 (SEP-856) is a novel psychotropic agent currently under phase III clinical investigation for schizophrenia treatment. In this study, we investigated the ability of an acute oral SEP-856 administration to modulate the functional activity of specific brain regions at basal levels and under glutamatergic or dopaminergic-perturbed conditions in adult rats. We found that immediate-early genes (IEGs) expression was strongly upregulated in the prefrontal cortex and, to a less extent, in the ventral hippocampus, suggesting an activation of these regions. Furthermore, SEP-856 was effective in preventing the hyperactivity induced by an acute injection of phencyclidine (PCP), but not of d-amphetamine (AMPH). The compound effectively normalized the PCP-induced increase in IEGs expression in the PFC at all doses tested, whereas only the highest dose determined the major modulations on AMPH-induced changes. Lastly, SEP-856 acute administration corrected the cognitive deficits produced by subchronic PCP administration. Taken together, our data provide further insights on SEP-856, suggesting that modulation of the PFC may represent an important mechanism for the functional and behavioural activity of this novel compound.


Asunto(s)
Antipsicóticos/farmacología , Cognición , Genes Inmediatos-Precoces , Piranos/farmacología , Esquizofrenia/tratamiento farmacológico , Administración Oral , Animales , Antipsicóticos/administración & dosificación , Antipsicóticos/uso terapéutico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Piranos/administración & dosificación , Piranos/uso terapéutico , Ratas , Ratas Sprague-Dawley
2.
Neurobiol Stress ; 13: 100271, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33344724

RESUMEN

Exposure to early life stress can interfere with neurodevelopmental trajectories to increase the vulnerability for psychiatric disorders later in life. With this respect, epigenetic mechanisms play a key role for the long-lasting changes in brain functions that may elicit and sustain psychopathologic outcomes. Here, we investigated DNA methylation changes as possible epigenetic mechanism mediating the effect of prenatal stress (PNS), an experimental paradigm associated with behavioral and molecular alterations relevant for psychiatric disorders. We identified 138 genes as being differentially methylated in the prefrontal cortex (PFC) and in the hippocampus (HIP) of male and female adult rats exposed to PNS. Among these genes, miR-30a and Neurod1 emerged as potential players for the negative outcomes associated with PNS exposure. Indeed, in addition to showing consistent methylation differences in both brain regions and in both sexes, and interacting with each other, they are both involved in Axon guidance and Neurotrophin signaling, which are important to neurodevelopmental disorders. We also found a significant reduction in the expression of a panel of genes (CAMK2A, c-JUN, LIMK1, MAP2K1, MAP2K2, PIK3CA and PLCG1) that belong to these two biological pathways and are also validated targets of miR-30a, pointing to a down-regulation of these pathways as a consequence of PNS exposure. Interestingly, we also found that miR-30a levels were significantly upregulated in depressed patients exposed to childhood trauma, as compared to control individuals. Importantly, we also found that a sub-chronic treatment with the atypical antipsychotic drug, lurasidone, during adolescence was able to prevent the up-regulation of miR-30a and normalized the expression of its target genes in response to PNS exposure. Our results demonstrate that miR-30a undergoes epigenetic changes following early life stress exposure and suggest that this miRNA could play a key role in producing broad and long-lasting alterations in neuroplasticity-related pathways, contributing to the etiology of psychiatric disorders.

3.
Mol Neurobiol ; 56(9): 6251-6260, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30746638

RESUMEN

Exposure to adverse events during gestation has detrimental effects on the maturation of specific brain networks, triggering changes in the expression of several stress-related mechanisms that may lead to long-lasting functional consequences, including cognitive deterioration. On these bases, the aim of the present study was to investigate the effects of early-life stress exposure on cognition and to explore potential molecular mechanisms contributing to the long-term functional impairment. We found that exposure to prenatal stress, a well-established animal model of early-life adversity, produces a significant disruption in the novel object recognition test both in male and female adult rats, although such impairment was more pronounced in females. Furthermore, the cognitive dysfunction observed during the behavioral test appears to be sustained by a disrupted activation of key networks of genes that may be required for proper cognitive performance. In particular, within the dorsal hippocampus, a brain region critical for cognition, the glucocorticoid, the inflammatory, and the protein kinase A signaling pathways are regulated by the novel object recognition test in an opposite manner in animals previously exposed to prenatal stress, when compared with control animals. These data further support the evidence that early-life stress exposure prompts cognitive impairment and suggest that this is the consequence of inability to activate the proper transcriptional machinery required for the cognitive performance.


Asunto(s)
Disfunción Cognitiva/genética , Efectos Tardíos de la Exposición Prenatal/genética , Estrés Fisiológico/genética , Transcripción Genética , Transcriptoma/genética , Animales , Disfunción Cognitiva/fisiopatología , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Memoria a Corto Plazo , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Transducción de Señal
4.
Transl Psychiatry ; 9(1): 23, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30655507

RESUMEN

Prenatal stress defines long-term phenotypes through epigenetic programming of the offspring. These effects are potentially mediated by glucocorticoid release and by sex. We hypothesized that the glucocorticoid receptor (Gr, Nr3c1) fashions the DNA methylation profile of offspring. Consistent with this hypothesis, fetal Nr3c1 heterozygosity leads to altered DNA methylation landscape in fetal placenta in a sex-specific manner. There was a significant overlap of differentially methylated genes in fetal placenta and adult frontal cortex in Nr3c1 heterozygotes. Phenotypically, Nr3c1 heterozygotes show significantly more anxiety-like behavior than wildtype. DNA methylation status of fetal placental tissue is significantly correlated with anxiety-like behavior of the same animals in adulthood. Thus, placental DNA methylation might predict behavioral phenotypes in adulthood. Our data supports the hypothesis that Nr3c1 influences DNA methylation at birth and that DNA methylation in placenta correlates with adult frontal cortex DNA methylation and anxiety-like phenotypes.


Asunto(s)
Trastornos de Ansiedad/genética , Conducta Animal , Metilación de ADN , Placenta , Receptores de Glucocorticoides/deficiencia , Factores Sexuales , Animales , Islas de CpG , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Feto , Masculino , Ratones , Ratones Noqueados , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética
5.
Nat Commun ; 9(1): 1596, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29686286

RESUMEN

Anorexia nervosa (AN) is a devastating eating disorder characterized by self-starvation that mainly affects women. Its etiology is unknown, which impedes successful treatment options leading to a limited chance of full recovery. Here, we show that gestation is a vulnerable window that can influence the predisposition to AN. By screening placental microRNA expression of naive and prenatally stressed (PNS) fetuses and assessing vulnerability to activity-based anorexia (ABA), we identify miR-340 as a sexually dimorphic regulator involved in prenatal programming of ABA. PNS caused gene-body hypermethylation of placental miR-340, which is associated with reduced miR-340 expression and increased protein levels of several target transcripts, GR, Cry2 and H3F3b. MiR-340 is linked to the expression of several nutrient transporters both in mice and human placentas. Using placenta-specific lentiviral transgenes and embryo transfer, we demonstrate the key role miR-340 plays in the mechanism involved in early life programming of ABA.


Asunto(s)
Anorexia Nerviosa/genética , MicroARNs/metabolismo , Placenta/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Adulto , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transferencia de Embrión , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , MicroARNs/genética , Actividad Motora , Embarazo , Análisis de Secuencia de ARN , Factores Sexuales
6.
Mol Psychiatry ; 23(11): 2192-2208, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29302075

RESUMEN

To date, gene-environment (GxE) interaction studies in depression have been limited to hypothesis-based candidate genes, since genome-wide (GWAS)-based GxE interaction studies would require enormous datasets with genetics, environmental, and clinical variables. We used a novel, cross-species and cross-tissues "omics" approach to identify genes predicting depression in response to stress in GxE interactions. We integrated the transcriptome and miRNome profiles from the hippocampus of adult rats exposed to prenatal stress (PNS) with transcriptome data obtained from blood mRNA of adult humans exposed to early life trauma, using a stringent statistical analyses pathway. Network analysis of the integrated gene lists identified the Forkhead box protein O1 (FoxO1), Alpha-2-Macroglobulin (A2M), and Transforming Growth Factor Beta 1 (TGF-ß1) as candidates to be tested for GxE interactions, in two GWAS samples of adults either with a range of childhood traumatic experiences (Grady Study Project, Atlanta, USA) or with separation from parents in childhood only (Helsinki Birth Cohort Study, Finland). After correction for multiple testing, a meta-analysis across both samples confirmed six FoxO1 SNPs showing significant GxE interactions with early life emotional stress in predicting depressive symptoms. Moreover, in vitro experiments in a human hippocampal progenitor cell line confirmed a functional role of FoxO1 in stress responsivity. In secondary analyses, A2M and TGF-ß1 showed significant GxE interactions with emotional, physical, and sexual abuse in the Grady Study. We therefore provide a successful 'hypothesis-free' approach for the identification and prioritization of candidate genes for GxE interaction studies that can be investigated in GWAS datasets.


Asunto(s)
Depresión/genética , Trastorno Depresivo/genética , Pruebas Genéticas/métodos , Adulto , Animales , Estudios de Cohortes , Depresión/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Humanos , Masculino , MicroARNs/análisis , MicroARNs/genética , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple/genética , Embarazo , Ratas , Transcriptoma/genética , Factor de Crecimiento Transformador beta1/genética , alfa-Macroglobulinas/genética , alfa-Macroglobulinas/metabolismo
7.
Neurosci Biobehav Rev ; 84: 352-358, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28843752

RESUMEN

A growing body of evidence supports the idea that drugs targeting the glutamate system may represent a valuable therapeutic alternative in major depressive disorders (MDD). The rapid and prolonged mood elevating effect of the NMDA receptor (NMDAR) antagonist ketamine has been studied intensely. However, its clinical use is hampered by deleterious side-effects, such as psychosis. Therefore, a better understanding of the mechanisms of the psychotropic effects after NMDAR blockade is necessary to develop glutamatergic antidepressants with improved therapeutic profile. Here we review recent experimental data that addressed molecular/cellular determinants of the antidepressant effect mediated by inactivating NMDAR subtypes. We refer to results obtained both in pharmacological and genetic animal models, ranging from global to conditional NMDAR manipulation. Our main focus is on the contribution of different NMDAR subtypes to the psychoactive effects induced by NMDAR ablation/blockade. We review data analyzing the effect of NMDAR subtype deletions limited to specific neuronal populations/brain areas in the regulation of mood. Altogether, these studies suggest effective and putative specific NMDAR drug targets for MDD treatment.


Asunto(s)
Antidepresivos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Terapia Molecular Dirigida/métodos , Isoformas de Proteínas/efectos de los fármacos , Animales , Humanos
8.
Eur Arch Psychiatry Clin Neurosci ; 268(1): 77-87, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27581816

RESUMEN

The evidence underlying the so-called glutamatergic hypothesis ranges from NMDA receptor hypofunction to an imbalance between excitatory and inhibitory circuits in specific brain structures. Among all glutamatergic system components, metabotropic receptors play a main role in regulating neuronal excitability and synaptic plasticity. Here, we investigated, using qRT-PCR and western blot, consequences in the hippocampus and prefrontal/frontal cortex (PFC/FC) of mice with a genetic deletion of the metabotropic glutamate receptor 5 (mGlu5), addressing key components of the GABAergic and glutamatergic systems. We found that mGlu5 knockout (KO) mice showed a significant reduction of reelin, GAD65, GAD67 and parvalbumin mRNA levels, which is specific for the PFC/FC, and that is paralleled by a significant reduction of protein levels in male KO mice. We next analyzed the main NMDA and AMPA receptor subunits, namely GluN1, GluN2A, GluN2B and GluA1, and we found that mGlu5 deletion determined a significant reduction of their mRNA levels, also within the hippocampus, with differences between the two genders. Our data suggest that neurochemical abnormalities impinging the glutamatergic and GABAergic systems may be responsible for the behavioral phenotype associated with mGlu5 KO animals and point to the close interaction of these molecular players for the development of neuropsychiatric disorders such as schizophrenia. These data could contribute to a better understanding of the involvement of mGlu5 alterations in the molecular imbalance between excitation and inhibition underlying the emergence of a schizophrenic-like phenotype and to understand the potential of mGlu5 modulators in reversing the deficits characterizing the schizophrenic pathology.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/genética , Receptor del Glutamato Metabotropico 5/deficiencia , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Parvalbúminas/genética , Parvalbúminas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptores Ionotrópicos de Glutamato/genética , Receptores Ionotrópicos de Glutamato/metabolismo , Proteína Reelina , Esquizofrenia/genética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Factores Sexuales
9.
Front Behav Neurosci ; 11: 113, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28706476

RESUMEN

Prenatal stress (PNS) might affect the developmental programming of adult chronic diseases such as metabolic and mood disorders. The molecular mechanisms underlying such regulations may rely upon long-term changes in stress-responsive effectors such as Brain-Derived Neurotrophic Factor (BDNF) that can affect neuronal plasticity underlying mood disorders and may also play a role in metabolic regulation. Based upon previous data, we hypothesized that PNS might lead to greater vulnerability to an obesogenic challenge experienced at adulthood. In order to investigate our hypothesis, pregnant Sprague-Dawley female rats underwent a chronic procedure of restraint stress during the last week of gestation. The adult offspring were then challenged with a high fat diet (HFD) over 8 weeks and tested for metabolic and emotional endpoints. Moreover, brain specific changes in Bdnf expression levels were also assessed. Overall, HFD resulted in increased caloric intake, insulin resistance, impaired glucose tolerance and higher circulating levels of leptin, while PNS increased the leptin/adiponectin ratio, an index of metabolic risk in adult male subjects. Interestingly, HFD consumption increased anxiety-like behaviors in the Elevated Plus Maze, particularly in males, and this effect was buffered by PNS. Levels of Bdnf were finely modulated by PNS and HFD in a region- and sex-dependent fashion: female offspring overall showed greater plasticity, possibly mediated through increased total Bdnf mRNA expression both in the hippocampus and in the hypothalamus. In conclusion, while the experience of maternal stress during intrauterine life promotes metabolic dysfunction induced by a HFD at adulthood, the interaction between PNS and HFD is positive in male subjects, and in agreement with the match-mismatch hypothesis, resulting in a reduction of anxious behaviors.

10.
Cereb Cortex ; 27(6): 3397-3413, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27797829

RESUMEN

Prenatal exposure to maternal infection increases the risk of neurodevelopmental disorders, including schizophrenia and autism. The molecular processes underlying this pathological association, however, are only partially understood. Here, we combined unbiased genome-wide transcriptional profiling with follow-up epigenetic analyses and structural magnetic resonance imaging to explore convergent molecular and neuromorphological alterations in corticostriatal areas of adult offspring exposed to prenatal immune activation. Genome-wide transcriptional profiling revealed that prenatal immune activation caused a differential expression of 116 and 251 genes in the medial prefrontal cortex and nucleus accumbens, respectively. A large part of genes that were commonly affected in both brain areas were related to myelin functionality and stability. Subsequent epigenetic analyses indicated that altered DNA methylation of promoter regions might contribute to the differential expression of myelin-related genes. Quantitative relaxometry comparing T1, T2, and myelin water fraction revealed sparse increases in T1 relaxation times and consistent reductions in T2 relaxation times. Together, our multi-system approach demonstrates that prenatal viral-like immune activation causes myelin-related transcriptional and epigenetic changes in corticostriatal areas. Even though these abnormalities do not seem to be associated with overt white matter reduction, they may provide a molecular mechanism whereby prenatal infection can impair myelin functionality and stability.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Imagen por Resonancia Magnética , Trastornos del Neurodesarrollo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Estudios de Cohortes , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Edad Gestacional , Inductores de Interferón/toxicidad , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Proteínas de la Mielina/metabolismo , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Poli I-C/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Reconocimiento en Psicología/efectos de los fármacos , Reconocimiento en Psicología/fisiología
11.
Pharmacol Ther ; 167: 13-27, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27452338

RESUMEN

The emergence of psychiatric disorders relies on the interaction between genetic vulnerability and environmental adversities. Several studies have demonstrated a crucial role for epigenetics (e.g. DNA methylation, post-translational histone modifications and microRNA-mediated post-transcriptional regulation) in the translation of environmental cues into adult behavioural outcome, which can prove to be harmful thus increasing the risk to develop psychopathology. Within this frame, non-coding RNAs, especially microRNAs, came to light as pivotal regulators of many biological processes occurring in the Central Nervous System, both during the neuronal development as well as in the regulation of adult function, including learning, memory and neuronal plasticity. On these basis, in recent years it has been hypothesised a central role for microRNA modulation and expression regulation in many brain disorders, including neurodegenerative disorders and mental illnesses. Indeed, the aim of the present review is to present the most recent state of the art regarding microRNA involvement in psychiatric disorders. We will first describe the mechanisms that regulate microRNA biogenesis and we will report evidences of microRNA dysregulation in peripheral body fluids, in postmortem brain tissues from patients suffering from psychopathology as well as in animal models. Last, we will discuss the potential to consider microRNAs as putative target for pharmacological intervention, using common psychotropic drugs or more specific tools, with the aim to normalize functions that are disrupted in different psychiatric conditions.


Asunto(s)
Trastornos Mentales/tratamiento farmacológico , MicroARNs/genética , Psicotrópicos/farmacología , Adulto , Animales , Encefalopatías/tratamiento farmacológico , Encefalopatías/genética , Encefalopatías/fisiopatología , Epigénesis Genética , Regulación de la Expresión Génica/genética , Humanos , Trastornos Mentales/genética , Trastornos Mentales/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología
12.
Dev Psychobiol ; 57(3): 365-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25783782

RESUMEN

Prenatal stress (PNS) is a risk factor for the development of neuropsychiatric disorders. This study was aimed at assessing, in a rodent model, changes in gene expression profiles and behavioral output as a result of PNS, during periadolescence, a critical developmental period for the onset of psychopathology. Social behavior was studied in a standardized social interaction paradigm and the expression of Brain-Derived Neurotrophic Factor (Bdnf), a marker of neuronal plasticity, and of inhibitory and excitatory mechanisms (Na(+)-K(+)-2Cl(-) and K(+)-Cl(-) cotransporters ratio, NKCC1/KCC2) was analyzed. Results indicate that PNS reduced Bdnf transcripts while increasing the NKCC1/KCC2 ratio, primarily in the hippocampus. In the prefrontal cortex, changes in Bdnf were found to be gender-dependent. These effects were accompanied by reduced levels of affiliative and investigative social behaviors. Interestingly, interaction with non-stressed subjects was able to improve sociality in PNS rats suggesting that the social environment could be exploited for therapeutic intervention.


Asunto(s)
Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Efectos Tardíos de la Exposición Prenatal/genética , Conducta Social , Estrés Fisiológico/genética , Estrés Psicológico/genética , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Femenino , Expresión Génica/genética , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Estrés Psicológico/fisiopatología
13.
Curr Pharm Des ; 21(11): 1388-95, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25564386

RESUMEN

The impact of early physical and social environments on life-long pathological phenotypes is well known and there is now compelling evidence that stressful experiences during gestation or early in life can lead to enhanced susceptibility to mental illness. Here, we discuss the data from preclinical studies aimed at investigating the molecular consequences of the exposure to stressful events during prenatal or early postnatal life that might contribute to later psychopathology. Particularly, we will discuss the existence of age windows of vulnerability to environmental conditions during brain maturation using as examples several studies performed with different animal models. Specifically, major deviations from normative neurobehavioural trajectories have been reported in animal models obtained following exposure to severe stress (maternal separation) ea rly in infancy or with rodent models of difficult and/or stressful pregnancies, including obstetric complications (e.g. prenatal restrain stress) and gestational exposure to infection (e.g prenatal immune challenge). These models have been associated with profound long-lasting deficits in the offspring's emotional and social behaviour, and with molecular changes associated with neuroplasticity.


Asunto(s)
Trastornos Mentales/etiología , Plasticidad Neuronal/fisiología , Estrés Psicológico/complicaciones , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Trastornos Mentales/epidemiología , Trastornos Mentales/fisiopatología , Embarazo , Complicaciones del Embarazo/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Factores de Tiempo
14.
Front Behav Neurosci ; 8: 407, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25505876

RESUMEN

The cyclic AMP (cAMP)-response element binding protein (CREB) is an activity-dependent transcription factor playing a role in synaptic plasticity, learning and memory, and emotional behavior. However, the impact of Creb ablation on rodent behavior is vague as e.g., memory performance of different Creb mutant mice depends on the specific type of mutation per se but additionally on the background and learning protocol differences. Here we present the first targeted ablation of CREB induced during adulthood selectively in principal forebrain neurons in a pure background strain of C57BL/6 mice. All hippocampal principal neurons exhibited lack of CREB expression. Mutant mice showed a severe anxiety phenotype in the openfield and novel object exploration test as well as in the Dark-Light Box Test, but unaltered hippocampus-dependent long-term memory in the Morris water maze and in context dependent fear conditioning. On the molecular level, CREB ablation led to CREM up regulation in the hippocampus and frontal cortex which may at least in part compensate for the loss of CREB. BDNF, a postulated CREB target gene, was down regulated in the frontal lobe but not in the hippocampus; neurogenesis remained unaltered. Our data indicate that in the adult mouse forebrain the late onset of CREB ablation can, in case of memory functionality, be compensated for and is not essential for memory consolidation and retrieval during adulthood. In contrast, the presence of CREB protein during adulthood seems to be pivotal for the regulation of emotional behavior.

15.
Int J Neuropsychopharmacol ; 18(4)2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25522402

RESUMEN

BACKGROUND: Major depression is associated with several alterations, including reduced neuronal plasticity and impaired synaptic function, which represent an important target of pharmacological intervention. METHODS: In the present study, we have investigated the ability of the antipsychotic drug lurasidone to modulate behavioral and neuroplastic alterations in the chronic mild stress model of depression. RESULTS: Rats that show reduced sucrose consumption after 2 weeks of chronic mild stress have reduced expression of the pool of Bdnf transcripts with the long 3' untranslated region (3'-UTR) that may be targeted to the synaptic compartment, suggesting the contribution of the neurotrophin to the behavioral dysfunction produced by chronic mild stress. The downregulation of Bdnf expression persisted also after 7 weeks of chronic mild stress, whereas chronic lurasidone treatment improved anhedonia in chronic mild stress rats and restored Bdnf mRNA levels in the prefrontal cortex. Moreover, chronic lurasidone treatment was able to normalize chronic mild stress-induced defects of Psd95 and Gfap as well as changes in molecular regulators of protein translation at the synapse, including mTOR and eEF2. CONCLUSIONS: These results demonstrate that lurasidone shows antidepressant properties in the chronic mild stress model through the modulation of synaptic and neuroplastic proteins. Such changes may contribute to the amelioration of functional capacities, which are deteriorated in patients with major depression and stress-related disorders.


Asunto(s)
Antidepresivos/farmacología , Trastorno Depresivo/tratamiento farmacológico , Clorhidrato de Lurasidona/farmacología , Corteza Prefrontal/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedad Crónica , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Quinasa del Factor 2 de Elongación/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 3 de Aminoácidos Excitadores/metabolismo , Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Wistar , Estrés Psicológico , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
16.
Hippocampus ; 24(4): 424-35, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24339333

RESUMEN

Adolescence is characterized by important molecular and anatomical changes with relevance for the maturation of brain circuitry and cognitive function. This time period is of critical importance in the emergence of several neuropsychiatric disorders accompanied by cognitive impairment, such as affective disorders and schizophrenia. The molecular mechanisms underlying these changes at neuronal level during this specific developmental stage remains however poorly understood. GluA1-containing AMPA receptors, which are located predominantly on hippocampal neurons, are the primary molecular determinants of synaptic plasticity. We investigated here the consequences of the inducible deletion of GluA1 AMPA receptors in glutamatergic neurons during late adolescence. We generated mutant mice with a tamoxifen-inducible deletion of GluA1 under the control of the CamKII promoter for temporally and spatially restricted gene manipulation. GluA1 ablation during late adolescence induced cognitive impairments, but also marked hyperlocomotion and sensorimotor gating deficits. Unlike the global genetic deletion of GluA1, inducible GluA1 ablation during late adolescence resulted in normal sociability. Deletion of GluA1 induced redistribution of GluA2 subunits, suggesting AMPA receptor trafficking deficits. Mutant animals showed increased hippocampal NMDA receptor expression and no change in striatal dopamine concentration. Our data provide new insight into the role of deficient AMPA receptors specifically during late adolescence in inducing several cognitive and behavioral alterations with possible relevance for neuropsychiatric disorders.


Asunto(s)
Trastornos del Conocimiento/metabolismo , Cuerpo Estriado/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Receptores AMPA/metabolismo , Conducta Social , Animales , Cuerpo Estriado/crecimiento & desarrollo , Dopamina/metabolismo , Hipocampo/crecimiento & desarrollo , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo , Trastornos Mentales , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Fenotipo , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Filtrado Sensorial/fisiología
17.
Pharmacol Res ; 80: 1-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24309096

RESUMEN

Despite the rapid control of schizophrenic symptoms is due to the ability of antipsychotic drugs (APDs) to block D2 receptors in the mesolimbic pathway, it is now well-established that the therapeutic effects rely on adaptive mechanisms set in motion by their long-term administration. Such neuroplastic mechanisms depend on the pharmacological profile of the drug employed, with marked differences existing between first and second generation APDs. On these bases, the major accomplishment of this work was to investigate neuroadaptive changes set in motion by repeated treatment with aripiprazole, a novel APD that is unique for being a partial agonist at dopamine D2 receptors. Moreover, given that stress plays a critical role in the exacerbation of disease symptoms, we also investigated whether aripiprazole could influence the dynamic response of the brain to an acute challenge. We found that repeated aripiprazole treatment in rats regulates the expression of different markers of neuroplasticity such as Bdnf, Arc and Npas4 in a brain-region specific fashion; more importantly, the expression of these molecules was significantly up-regulated by an acute swim stress only in aripiprazole-treated animals, which is suggestive of increased ability to cope with the adverse event. We indeed found an overall facilitation of Bdnf expression, an effect that is mainly evident in the prefrontal cortex on the pool of transcripts undergoing dendritic localization. Overall, our results provide novel information regarding the mechanisms through which aripiprazole may regulate brain function and could contribute to improve neuroplastic defects that are associated with schizophrenia symptomatology.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Proteínas del Citoesqueleto/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Piperazinas/farmacología , Corteza Prefrontal/metabolismo , Quinolonas/farmacología , Estrés Fisiológico/efectos de los fármacos , Animales , Antipsicóticos/farmacología , Aripiprazol , Esquema de Medicación , Masculino , Piperazinas/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Quinolonas/administración & dosificación , Ratas
18.
Artículo en Inglés | MEDLINE | ID: mdl-24361635

RESUMEN

Lurasidone is a novel second generation antipsychotic drug characterized by a multi-receptor profile. Besides the high affinity for 5-HT2A and D2 receptors, it is also characterized by potent 5-HT7 receptor antagonism, which may be beneficial for mood and cognition. Considering that dose-dependent changes in receptor occupancy may differentially impact gene transcription, we aimed at investigating the effects of acute and chronic treatments with different doses of lurasidone (1, 3 and 10mg/kg) in rats on the expression of the activity-regulated genes Arc, Zif268 and Npas4, which are markers of neuronal activation and are also associated with neuroadaptive mechanisms. Our results show dose-dependent and anatomically-selective differences after acute and chronic lurasidone treatment. Indeed, the effects produced by acute treatment seem to reflect the modulatory activity of lurasidone at selected neurotransmitter receptors. In fact, low doses of the drug acted in the hippocampus, while high doses acted in the striatum, reflecting the high predominance of D2 receptor expression in this brain region. On the contrary, chronic treatment with lurasidone revealed a different profile of IEGs modulation, possibly reflecting neuroadaptive changes set in motion in response to repetitive drug exposure. In summary, the multi-receptor profile of lurasidone leads to the recruitment of different brain structures in a dose-related manner and this may be important for its therapeutic properties, particularly with respect to antidepressant activity and cognition.


Asunto(s)
Antipsicóticos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Proteínas del Citoesqueleto/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Isoindoles/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Tiazoles/farmacología , Animales , Antipsicóticos/administración & dosificación , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Isoindoles/administración & dosificación , Clorhidrato de Lurasidona , Masculino , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Tiazoles/administración & dosificación
19.
Proc Natl Acad Sci U S A ; 110(21): 8708-13, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23650397

RESUMEN

Stress and glucocorticoid hormones regulate hippocampal neurogenesis, but the molecular mechanisms mediating these effects are poorly understood. Here we identify the glucocorticoid receptor (GR) target gene, serum- and glucocorticoid-inducible kinase 1 (SGK1), as one such mechanism. Using a human hippocampal progenitor cell line, we found that a small molecule inhibitor for SGK1, GSK650394, counteracted the cortisol-induced reduction in neurogenesis. Moreover, gene expression and pathway analysis showed that inhibition of the neurogenic Hedgehog pathway by cortisol was SGK1-dependent. SGK1 also potentiated and maintained GR activation in the presence of cortisol, and even after cortisol withdrawal, by increasing GR phosphorylation and GR nuclear translocation. Experiments combining the inhibitor for SGK1, GSK650394, with the GR antagonist, RU486, demonstrated that SGK1 was involved in the cortisol-induced reduction in progenitor proliferation both downstream of GR, by regulating relevant target genes, and upstream of GR, by increasing GR function. Corroborating the relevance of these findings in clinical and rodent settings, we also observed a significant increase of SGK1 mRNA in peripheral blood of drug-free depressed patients, as well as in the hippocampus of rats subjected to either unpredictable chronic mild stress or prenatal stress. Our findings identify SGK1 as a mediator for the effects of cortisol on neurogenesis and GR function, with particular relevance to stress and depression.


Asunto(s)
Depresión/enzimología , Glucocorticoides/metabolismo , Hipocampo/enzimología , Hidrocortisona/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Glucocorticoides/metabolismo , Estrés Fisiológico , Transporte Activo de Núcleo Celular/efectos de los fármacos , Adulto , Animales , Benzoatos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Transformada , Núcleo Celular/metabolismo , Núcleo Celular/patología , Depresión/patología , Femenino , Proteínas Hedgehog/metabolismo , Hipocampo/patología , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Masculino , Persona de Mediana Edad , Mifepristona/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
20.
Neuropsychopharmacology ; 38(5): 872-83, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23303060

RESUMEN

Stress and glucocorticoid hormones regulate hippocampal neurogenesis, but the molecular mechanisms underlying their effects are unknown. We, therefore, investigated the molecular signaling pathways mediating the effects of cortisol on proliferation, neuronal differentiation, and astrogliogenesis, in an immortalized human hippocampal progenitor cell line. In addition, we examined the molecular signaling pathways activated in the hippocampus of prenatally stressed rats, characterized by persistently elevated glucocorticoid levels in adulthood. In human hippocampal progenitor cells, we found that low concentrations of cortisol (100 nM) increased proliferation (+16%), decreased neurogenesis into microtubule-associated protein 2 (MAP2)-positive neurons (-24%) and doublecortin (Dcx)-positive neuroblasts (-21%), and increased differentiation into S100ß-positive astrocytes (+23%). These effects were dependent on the mineralocorticoid receptor (MR) as they were abolished by the MR antagonist, spironolactone, and mimicked by the MR-agonist, aldosterone. In contrast, high concentrations of cortisol (100 µM) decreased proliferation (-17%) and neuronal differentiation into MAP2-positive neurons (-22%) and into Dcx-positive neuroblasts (-27%), without regulating astrogliogenesis. These effects were dependent on the glucocorticoid receptor (GR), blocked by the GR antagonist RU486, and mimicked by the GR-agonist, dexamethasone. Gene expression microarray and pathway analysis showed that the low concentration of cortisol enhances Notch/Hes-signaling, the high concentration inhibits TGFß-SMAD2/3-signaling, and both concentrations inhibit Hedgehog signaling. Mechanistically, we show that reduced Hedgehog signaling indeed critically contributes to the cortisol-induced reduction in neuronal differentiation. Accordingly, TGFß-SMAD2/3 and Hedgehog signaling were also inhibited in the hippocampus of adult prenatally stressed rats with high glucocorticoid levels. In conclusion, our data demonstrate novel molecular signaling pathways that are regulated by glucocorticoids in vitro, in human hippocampal progenitor cells, and by stress in vivo, in the rat hippocampus.


Asunto(s)
Glucocorticoides/metabolismo , Hipocampo/citología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Transducción de Señal/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Femenino , Feto , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Erizos/metabolismo , Antagonistas de Hormonas/farmacología , Humanos , Hidrocortisona/farmacología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Mifepristona/farmacología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...