Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
1.
Neuropsychopharmacology ; 47(3): 687-695, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34743200

RESUMEN

Hippocampal hyperactivity driven by GABAergic interneuron deficits and NMDA receptor hypofunction is associated with the hyperdopaminergic state often observed in schizophrenia. Furthermore, previous research in the methylazoxymethanol acetate (MAM) rat model has demonstrated that repeated peripubertal diazepam administration can prevent the emergence of adult hippocampal hyperactivity, dopamine-system hyperactivity, and associated psychosis-relevant behaviors. Here, we sought to characterize hippocampal GABAA and NMDA receptors in MAM-treated rats and to elucidate the receptor mechanisms underlying the promising effects of peripubertal diazepam exposure. Quantitative receptor autoradiography was used to measure receptor density in the dorsal hippocampus CA1, ventral hippocampus CA1, and ventral subiculum. Specifically, [3H]-Ro15-4513 was used to quantify the density of α5GABAA receptors (α5GABAAR), [3H]-flumazenil to quantify α1-3;5GABAAR, and [3H]-MK801 to quantify NMDA receptors. MAM rats exhibited anxiety and schizophrenia-relevant behaviors as measured by elevated plus maze and amphetamine-induced hyperlocomotion (AIH), although diazepam only partially rescued these behaviors. α5GABAAR density was reduced in MAM-treated rats in all hippocampal sub-regions, and negatively correlated with AIH. Ventral hippocampus CA1 α5GABAAR density was positively correlated with anxiety-like behavior. Dorsal hippocampus CA1 NMDA receptor density was increased in MAM-treated rats, and positively correlated with AIH. [3H]-flumazenil revealed no significant effects. Finally, we found no significant effect of diazepam treatment on receptor densities, potentially related to the only partial rescue of schizophrenia-relevant phenotypes. Overall, our findings provide first evidence of α5GABAAR and NMDA receptor abnormalities in the MAM model, suggesting that more selective pharmacological agents may become a novel therapeutic mechanism in schizophrenia.


Asunto(s)
Acetato de Metilazoximetanol , Esquizofrenia , Animales , Modelos Animales de Enfermedad , Hipocampo , Acetato de Metilazoximetanol/farmacología , Ratas , Receptores de N-Metil-D-Aspartato , Ácido gamma-Aminobutírico
2.
Mol Psychiatry ; 26(12): 7679-7689, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34193975

RESUMEN

Recent evidence showed thalamic abnormalities in schizophrenia involving disruptions to the parvalbumin neurons in the thalamic reticular nucleus (TRN). However, their functional consequences, as well as a potential linkage to oxidative stress, are unclear. The TRN is posited to gate prefrontal control of dopamine neuron activity in the ventral tegmental area (VTA). Thus, we hypothesized that schizophrenia-related TRN abnormalities might contribute to dopamine dysregulation, a well-known feature of the disorder. To test this, in adult rats exposed prenatally to methylazoxymethanol acetate (MAM rats), oxidative impairments to the parvalbumin neurons in the anterior TRN were assessed by immunohistochemistry. Using in vivo electrophysiology, we investigated whether inactivation of the prefrontal cortex would produce differential effects on VTA dopamine neurons in MAM rats. We show that MAM rats displayed reduced markers of parvalbumin and wisteria floribunda agglutinin-labeled perineuronal nets, correlating with increased markers of oxidative stress (8-oxo-7, 8-dihydro-20-deoxyguanosine, and 3-nitrotyrosine). Moreover, MAM rats displayed heightened baseline and abnormal prefrontal control of VTA dopamine neuron activity, as tetrodotoxin-induced inactivation of the infralimbic prefrontal cortex decreased the dopamine population activity, contrary to the normal increase in controls. Such dopamine neuron dysregulation was recapitulated by enzymatic perineuronal net digestion in the TRN of normal rats. Furthermore, juvenile (postnatal day 11-25) antioxidant treatment (N-acetyl-cysteine, 900 mg/L drinking water) prevented all these impairments in MAM rats. Our findings suggest that early accumulation of oxidative stress in the TRN may shape the later onset of schizophrenia pathophysiology, highlighting redox regulation as a potential target for early intervention.


Asunto(s)
Dopamina , Esquizofrenia , Acetilcisteína/farmacología , Animales , Modelos Animales de Enfermedad , Dopamina/farmacología , Neuronas Dopaminérgicas/fisiología , Acetato de Metilazoximetanol/farmacología , Ratas , Núcleos Talámicos
3.
Behav Brain Res ; 412: 113442, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34229023

RESUMEN

Malformation of cortical developments (MCDs) is currently an incurable disease and is associated with significant neuropsychological problems, such as intellectual disability, epilepsy, and anxiety disorders from a young age. Development of a suitable animal model and pathophysiological study is therefore necessary to better understand and treat MCDs from being an incurable disease. The Y-maze, open field, and fear conditioning studies were performed at postnatal days 40-44 to validate the behavioral phenotypes of the existing rat model of MCD with prenatal methylazoxymethanol exposure at their developmental period. The study results show that juvenile rats with MCD spent significantly less time inside the novel arms in Y-maze and less time in the peripheral zones of the open field. Additionally, the rats with MCDs showed attenuated freezing behavior to sound and light cues as well as to context after fear conditioning. This comprehensive behavioral analysis of rats with MCDs at the juvenile period indicate a lack of spatial memory, decreased anxiety, and learning disability in these rats, which is compatible with the human behavioral phenotype of MCDs and can be used as the behavioral biomarkers for future translational research.


Asunto(s)
Malformaciones del Desarrollo Cortical/fisiopatología , Memoria a Corto Plazo/fisiología , Conducta de Reducción del Riesgo , Afecto , Animales , Ansiedad/fisiopatología , Conducta Animal/fisiología , Encéfalo/metabolismo , Cognición/fisiología , Señales (Psicología) , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Masculino , Malformaciones del Desarrollo Cortical/psicología , Aprendizaje por Laberinto/fisiología , Acetato de Metilazoximetanol/efectos adversos , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Actividad Motora/fisiología , Neurogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Asunción de Riesgos , Memoria Espacial/fisiología
4.
Schizophr Bull ; 47(6): 1806-1814, 2021 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33928393

RESUMEN

Pomaglumetad methionil (POM), a group 2 metabotropic glutamate receptor (mGluR2/3) agonist, showed promise as a novel antipsychotic in preclinical research but failed to show efficacy in clinical trials, though it has been suggested that it may be effective in certain patient populations, including early in disease patients. We used the methyazoxymethanol acetate (MAM) rat model of schizophrenia to determine whether POM may prevent the development of dopamine (DA) system dysfunction in a model representative of the hyperdopaminergic state thought to underlie psychosis, compared to control (SAL) rats. MAM and SAL rats were administered either POM (3 mg/kg, i.p.), vehicle (1 ml/kg), or no injection during postnatal day (PD) 31-40. In either late adolescence (PD 47-56) or adulthood (PD 83-96), novel object recognition (NOR) was tested, followed by anesthetized in vivo electrophysiological recordings of VTA DA neuron activity or ventral hippocampal (vHPC) pyramidal neuron activity. MAM rats treated with POM demonstrated increased NOR in adulthood compared to no injection MAM rats, but not compared to vehicle-treated MAM rats. POM-treated MAM rats demonstrated normalized DA neuron population activity and vHPC pyramidal neuron activity compared to vehicle and no injection MAM rats in both late adolescence and adulthood. No significant differences were observed across treatment groups in SAL rats. These results suggest that peripubertal mGluR2/3 agonist administration can prevent the emergence of vHPC pyramidal neuron hyperactivity and increased DA neuron population activity in adult MAM rats.


Asunto(s)
Aminoácidos/farmacología , Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Esquizofrenia/tratamiento farmacológico , Área Tegmental Ventral/efectos de los fármacos , Factores de Edad , Aminoácidos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/administración & dosificación , Masculino , Acetato de Metilazoximetanol/farmacología , Neurotoxinas/farmacología , Ratas
5.
Behav Brain Res ; 406: 113231, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33737089

RESUMEN

Adolescent social isolation (SI) might change the trajectory of brain development. In the present study, we investigated the effect of short-term adolescent SI on fear memory, anxiety and protein levels in the adult medial prefrontal cortex of rats prenatally treated with methylazoxymethanol, MAM-E17 model of schizophrenia. The animals were maintained in standard housing (SH) or social isolation (P30-P40, SI) conditions. Behavioural tests (trace or delay fear conditioning, light/dark box) were performed in late adolescence and early adulthood. The results showed that MAM treatment did not alter fear memory, which was investigated with the use of either trace or delay fear conditioning, at any age, and SI decreased the fear response in adult control animals only under trace conditioning. Neither MAM nor SI influenced anxiety-related behaviour measured in the light/dark box. A proteomics study showed that both MAM and SI changed the protein levels related to synapse maturation and cytoskeletal organization, energy transfer and metabolic processes. Prenatal or adolescent environmental factors are able to change the expression of proteins that are correlated with behavioural impairments. Moreover, SI reversed some alterations in proteins induced by MAM. Thus, normally developing brains showed different responses to adolescent SI than those with altering courses of MAM administration.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Corteza Prefrontal , Efectos Tardíos de la Exposición Prenatal , Esquizofrenia , Aislamiento Social , Factores de Edad , Animales , Femenino , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Proteoma , Ratas Wistar , Esquizofrenia/etiología , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Teratógenos/farmacología
6.
Artículo en Inglés | MEDLINE | ID: mdl-33152383

RESUMEN

Schizophrenia is a severe neuropsychiatric disease associated with substantially higher mortality. Reduced life expectancy in schizophrenia relates to an increased prevalence of metabolic disturbance, and antipsychotic medication is a major contributor. Molecular mechanisms underlying adverse metabolic effects of antipsychotics are not fully understood; however, adipose tissue homeostasis deregulation appears to be a critical factor. We employed mass spectrometry-based untargeted proteomics to assess the effect of chronic olanzapine, risperidone, and haloperidol treatment in visceral adipose tissue of prenatally methylazoxymethanol (MAM) acetate exposed rats, a well-validated neurodevelopmental animal model of schizophrenia. Bioinformatics analysis of differentially expressed proteins was performed to highlight the pathways affected by MAM and the antipsychotics treatment. MAM model was associated with the deregulation of the TOR (target of rapamycin) signalling pathway. Notably, alterations in protein expression triggered by antipsychotics were observed only in schizophrenia-like MAM animals where we revealed hundreds of affected proteins according to our two-fold threshold, but not in control animals. Treatments with all antipsychotics in MAM rats resulted in the downregulation of mRNA processing and splicing, while drug-specific effects included among others upregulation of insulin resistance (olanzapine), upregulation of fatty acid metabolism (risperidone), and upregulation of nucleic acid metabolism (haloperidol). Our data indicate that deregulation of several energetic and metabolic pathways in adipose tissue is associated with APs administration and is prominent in MAM schizophrenia-like model but not in control animals.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Antipsicóticos/uso terapéutico , Grasa Intraabdominal/efectos de los fármacos , Acetato de Metilazoximetanol/farmacología , Esquizofrenia/tratamiento farmacológico , Tejido Adiposo/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Haloperidol/farmacología , Haloperidol/uso terapéutico , Grasa Intraabdominal/embriología , Grasa Intraabdominal/metabolismo , Olanzapina/farmacología , Olanzapina/uso terapéutico , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteómica , Ratas , Ratas Sprague-Dawley , Risperidona/farmacología , Risperidona/uso terapéutico , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
7.
PLoS One ; 15(6): e0232200, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32497066

RESUMEN

Schizophrenia is a debilitating disorder affecting just under 1% of the population. While the symptoms of this disorder do not appear until late adolescence, pathological alterations likely occur earlier, during development in utero. While there is an increasing literature examining transcriptome alterations in patients, it is not possible to examine the changes in gene expression that occur during development in humans that will develop schizophrenia. Here we utilize three distinct rodent developmental disruption models of schizophrenia to examine potential overlapping alterations in the transcriptome, with a specific focus on markers of interneuron development. Specifically, we administered either methylazoxymethanol acetate (MAM), Polyinosinic:polycytidylic acid (Poly I:C), or chronic protein malnutrition, on GD 17 and examined mRNA expression in the developing hippocampus of the offspring 18 hours later. Here, we report alterations in gene expression that may contribute to the pathophysiology of schizophrenia, including significant alterations in interneuron development and ribosome function.


Asunto(s)
Perfilación de la Expresión Génica , Crecimiento y Desarrollo , Esquizofrenia/genética , Animales , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Crecimiento y Desarrollo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Desnutrición/complicaciones , Acetato de Metilazoximetanol/farmacología , Poli I-C/farmacología , Embarazo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/etiología , Esquizofrenia/fisiopatología
8.
Schizophr Bull ; 46(3): 612-622, 2020 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-31738422

RESUMEN

Prenatal administration of mitotoxin methylazoxymethanol acetate (MAM) in rats produces behavioral, pharmacological, and anatomical abnormalities once offspring reach adulthood, thus establishing a widely used neurodevelopmental model of schizophrenia. However, the molecular aspects underlying this disease model are not well understood. Therefore, this study examines epigenetic and transcriptional dysregulation in the prefrontal cortex and hippocampus of MAM rats as these are brain regions closely associated with schizophrenia pathogenesis. Upon sequencing messenger and microRNA (mRNA and miRNA, respectively), differential expression was revealed in the prefrontal cortex and hippocampus between MAM- and saline-treated rats; sequencing data were validated by qualitative real-time polymerase chain reaction. Bioinformatic analyses demonstrated that the differentially expressed (DE) genes were strongly enriched in interactive pathways related to schizophrenia, including chemical synaptic transmission, cognition, and inflammatory responses; also, the potential target genes of the DE miRNAs were enriched in pathways related to synapses and inflammation. The blood of schizophrenia patients and healthy controls was further analyzed for several top DE mRNAs: DOPA decarboxylase, ret proto-oncogene, Fc receptor-like 2, interferon lambda receptor 1, and myxovirus (influenza virus) resistance 2. The results demonstrated that the expression of these genes was dysregulated in patients with schizophrenia; combining these mRNAs sufficiently differentiated schizophrenia patients from controls. Taken together, this study suggests that the MAM model has the potential to reproduce hippocampus and prefrontal cortex abnormalities, relevant to schizophrenia, at the epigenetic and transcriptional levels. These data also provide novel targets for schizophrenia diagnoses and treatments.


Asunto(s)
Epigénesis Genética , Hipocampo , Acetato de Metilazoximetanol/farmacología , Trastornos del Neurodesarrollo , Neurotoxinas/farmacología , Corteza Prefrontal , Esquizofrenia , Transcripción Genética , Adulto , Animales , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Masculino , MicroARNs , Trastornos del Neurodesarrollo/sangre , Trastornos del Neurodesarrollo/inducido químicamente , Trastornos del Neurodesarrollo/genética , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Embarazo , Proto-Oncogenes Mas , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Esquizofrenia/sangre , Esquizofrenia/inducido químicamente , Esquizofrenia/genética , Análisis de Secuencia de ARN , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Investigación Biomédica Traslacional
9.
Behav Brain Res ; 362: 258-265, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30660776

RESUMEN

Although there are clear sex differences in individuals with schizophrenia, preclinical research has historically favored the use of male rats for behavioral studies. The methylazoxymethanol acetate (MAM) model is a gestational disruption model of schizophrenia and has been reported to produce robust behavioral, neurophysiological and anatomical alterations in male rats; however, whether similar effects are observed in female rats is less well known. In this study, we characterize the behavioral, electrophysiological and molecular alterations induced by prenatal MAM administration in female rats while also examining the potential effects of the estrous cycle on schizophrenia-like behaviors. Specifically, MAM-treated female offspring demonstrated deficits in sensorimotor gating, latent inhibition, and social interaction, consistent with those observed in male animals. Interestingly, amphetamine-induced locomotor activity, latent inhibition, and social interaction were also affected by the estrous cycle. To examine the potential cellular mechanisms associated with these behavioral alterations, we analyzed hippocampal parvalbumin (PV) interneurons. Deficits in PV interneuron number and high-frequency gamma oscillations were disrupted in female MAM-treated rats regardless of the stage of the estrous cycle; however, alterations in PV protein expression were more prominent during metestrus/diestrus. Taken together, these data suggest that prenatal MAM exposure in female rats produces robust behavioral, molecular, and physiological deficits consistent with those observed in the male MAM model of schizophrenia. Moreover, our results also suggest that specific schizophrenia-like symptoms can also be influenced by the estrous cycle, and further emphasize the importance of sex as a biological variable when using preclinical models.


Asunto(s)
Conducta Animal/efectos de los fármacos , Ciclo Estral/fisiología , Acetato de Metilazoximetanol/farmacología , Esquizofrenia/fisiopatología , Filtrado Sensorial/efectos de los fármacos , Anfetamina/farmacología , Animales , Modelos Animales de Enfermedad , Ciclo Estral/efectos de los fármacos , Femenino , Hipocampo/metabolismo , Parvalbúminas/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Sprague-Dawley , Esquizofrenia/metabolismo
10.
Neuropharmacology ; 146: 212-221, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30496751

RESUMEN

In agreement with the neurodevelopmental hypothesis of schizophrenia, prenatal exposure of rats to the antimitotic agent methylazoxymethanol acetate (MAM) at gestational day 17 produced long-lasting behavioral alterations such as social withdrawal and cognitive impairment in the social interaction test and in the novel object recognition test, respectively. At the molecular level, an increased cannabinoid receptor type-1 (CB1) mRNA and protein expression, which might be due to reduction in DNA methylation at the gene promoter in the prefrontal cortex (PFC), coincided with deficits in the social interaction test and in the novel object recognition test in MAM rats. Both the schizophrenia-like phenotype and altered transcriptional regulation of CB1 receptors were reversed by peripubertal treatment (from PND 19 to PND 39) with the non-psychotropic phytocannabinoid cannabidiol (30 mg/kg/day), or, in part, by treatment with the cannabinoid CB1 receptor antagonist/inverse agonist AM251 (0.5 mg/kg/day), but not with haloperidol (0.6 mg/kg/day). These results suggest that early treatment with cannabidiol may prevent both the appearance of schizophrenia-like deficits as well as CB1 alterations in the PFC at adulthood, supporting that peripubertal cannabidiol treatment might be protective against MAM insult.


Asunto(s)
Cannabidiol/farmacología , Acetato de Metilazoximetanol/farmacología , Efectos Tardíos de la Exposición Prenatal/tratamiento farmacológico , Esquizofrenia/tratamiento farmacológico , Amidas , Animales , Ácidos Araquidónicos/metabolismo , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Etanolaminas/metabolismo , Femenino , Glicéridos/metabolismo , Hipocampo/metabolismo , Relaciones Interpersonales , Masculino , Actividad Motora/efectos de los fármacos , Ácidos Oléicos/metabolismo , Ácidos Palmíticos/metabolismo , Piperidinas/farmacología , Alcamidas Poliinsaturadas/metabolismo , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Pubertad , Pirazoles/farmacología , ARN Mensajero/metabolismo , Ratas , Receptor Cannabinoide CB1/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo
11.
Neuropharmacology ; 146: 28-38, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30448422

RESUMEN

In an animal neurodevelopmental model of schizophrenia, we investigated ultrasonic communication and social behavior in male and female rats. Pregnant dams were treated with methylazoxymethanol acetate (MAM; 22 mg/kg) at 17 days of gestation. First, we examined the ultrasonic vocalizations (USVs) emitted by 8-day-old pups isolated from their mothers and placed in a familiar or an unfamiliar environment. Second, we assessed tickling-induced USVs, social play (SP) behavior and accompanying USVs in 30-day-old juveniles. Independent of the prenatal treatment, sex differences were noted at both ages. In the pups isolated from their mothers, compared to the females, the males produced flatter calls with a lower frequency. Compared to the females, the tickling-induced male USVs were characterized by a higher frequency, and the male SP-induced USVs showed a broader bandwidth and more modulated structure. Additionally, the numbers of both SP-induced USVs and SP episodes in the males were higher than those in the females. In contrast, the MAM exposure reduced the ultrasonic communication and social behavior independent of age almost equally in the male and female rats. The MAM-exposed isolated pups and juveniles experiencing tickling and social interaction displayed lower USV bandwidths, suggesting that the complexity of their ultrasonic communication was reduced. In addition, the MAM-exposed juveniles demonstrated a lower number of 50-kHz "happy calls" and decreased SP duration, which is suggestive of social withdrawal or negative-like symptoms. These data demonstrate that young MAM-exposed rats display an atypical repertoire of USVs and reduced play behavior suggestive of communication deficits associated with schizophrenia.


Asunto(s)
Esquizofrenia/fisiopatología , Ultrasonido , Vocalización Animal/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Acetato de Metilazoximetanol/farmacología , Embarazo , Ratas , Esquizofrenia/inducido químicamente , Conducta Social
12.
Neuropsychopharmacology ; 44(3): 572-580, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30267014

RESUMEN

Aripiprazole is an antipsychotic drug characterized by partial agonist activity at D2 receptors to normalize both hyperdopaminergic and hypodopaminergic states. Traditional D2 antagonist antipsychotic drugs have been shown previously to reduce dopamine neuron activity through action on D2 autoreceptors to produce an overexcitation-induced cessation of cell firing, referred to as depolarization block. It is unclear whether aripiprazole reduces dopamine neuron activity via inhibition or, as seen following D2 antagonist administration, depolarization block. The impact of acute and repeated aripiprazole treatment was examined in the methylazoxymethanol acetate (MAM) rodent model to observe its effects on a hyperdopaminergic system, compared to normal rats. We found that administration of aripiprazole acutely or after 1 or 7 days of withdrawal from 21-day repeated treatment led to a decrease in the number of spontaneously active dopamine neurons in MAM rats but not in controls. This reduction was not reversed by apomorphine (100-200 µg/kg i.p. or 20 µg/kg i.v.) administration, suggesting that it was not due to depolarization block. In contrast, 1 h after induction of depolarization block of dopamine neurons by acute haloperidol treatment (0.6 mg/kg i.p.), aripiprazole (1 mg/kg, i.p.) reversed the depolarization block state. Therefore, aripiprazole rapidly reduced the hyperdopaminergic activity selectively in MAM rats. The reduction is unlikely due to depolarization block and persists following 7-day withdrawal from repeated treatment. Aripiprazole also removes haloperidol-induced depolarization block in MAM rats, which may underlie the acute psychotic state often observed with switching to this treatment.


Asunto(s)
Aripiprazol/farmacología , Agonistas de Dopamina/farmacología , Antagonistas de Dopamina/farmacología , Neuronas Dopaminérgicas/efectos de los fármacos , Receptores de Dopamina D2/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Animales , Apomorfina/farmacología , Modelos Animales de Enfermedad , Electroencefalografía , Haloperidol/farmacología , Masculino , Acetato de Metilazoximetanol/farmacología , Ratas , Ratas Sprague-Dawley
13.
Dev Neurobiol ; 78(12): 1206-1217, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30246945

RESUMEN

Adult hippocampal neurogenesis has been proposed to both aid memory formation and disrupt memory. We examined the role of adult hippocampal neurogenesis in spatial working and reference memory in black-capped chickadees (Poecile atricapillus), a passerine bird that relies on spatial memory for cache retrieval and foraging. We tested spatial working and spatial reference memory in birds that had received methylazoxymethanol acetate (MAM), a neurotoxin that decreases hippocampal neurogenesis. MAM treatment significantly reduced neurogenesis in the hippocampus quantified by doublecortin (DCX) labeling of newly divided and migrating neurons. MAM treatment had little effect on the working or reference memory but caused an increase in errors on the reference memory task following reversal. Working memory for recently visited spatial locations and reference memory for familiar spatial locations were thus unaffected by a reduction in neurogenesis. An increase in errors following reference memory reversal may indicate that adult hippocampal neurogenesis aids in pattern separation, the differentiation of similar memories at the time of encoding.


Asunto(s)
Conducta Animal/fisiología , Hipocampo/fisiología , Memoria a Corto Plazo/fisiología , Neurogénesis/fisiología , Aprendizaje Inverso/fisiología , Pájaros Cantores/fisiología , Memoria Espacial/fisiología , Animales , Conducta Animal/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Acetato de Metilazoximetanol/farmacología , Neurogénesis/efectos de los fármacos , Neurotoxinas/farmacología , Aprendizaje Inverso/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
14.
Neurobiol Learn Mem ; 134 Pt B: 294-303, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27485950

RESUMEN

Cognitive impairments are amongst the most debilitating deficits of schizophrenia and the best predictor of functional outcome. Schizophrenia is hypothesized to have a neurodevelopmental origin, making animal models of neurodevelopmental insult important for testing predictions that early insults will impair cognitive function. Rats exposed to methylazoxymethanol acetate (MAM) at gestational day 17 display morphological, physiological and behavioral abnormalities relevant to schizophrenia. Here we investigate the cognitive abilities of adult MAM rats. We examined brain activity in MAM rats by histochemically assessing cytochrome oxidase enzyme activity, a metabolic marker of neuronal activity. To assess cognition, we used a hippocampus-dependent two-frame active place avoidance paradigm to examine learning and spatial memory, as well as cognitive control and flexibility using the same environment and evaluating the same set of behaviors. We confirmed that adult MAM rats have altered hippocampal morphology and brain function, and that they are hyperactive in an open field. The latter likely indicates MAM rats have a sensorimotor gating deficit that is common to many animal models used for schizophrenia research. On first inspection, cognitive control seems impaired in MAM rats, indicated by more errors during the two-frame active place avoidance task. Because MAM rats are hyperactive throughout place avoidance training, we considered the possibility that the hyperlocomotion may account for the apparent cognitive deficits. These deficits were reduced on the basis of measures of cognitive performance that account for motor activity differences. However, though other aspects of memory are intact, the ability of MAM rats to express trial-to-trial memory is delayed compared to control rats. These findings suggest that spatial learning and cognitive abilities are largely intact, that the most prominent cognitive deficit is specific to acquiring memory in the MAM neurodevelopmental model, and that hyperactivity can confound assessments of cognition in animal models of mental dysfunction.


Asunto(s)
Función Ejecutiva/fisiología , Hipocampo/fisiopatología , Hipercinesia/fisiopatología , Trastornos de la Memoria/fisiopatología , Acetato de Metilazoximetanol/farmacología , Corteza Prefrontal/fisiopatología , Esquizofrenia/fisiopatología , Memoria Espacial/fisiología , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Masculino , Embarazo , Ratas , Ratas Long-Evans , Esquizofrenia/inducido químicamente
15.
Neuropsychopharmacology ; 41(13): 3003-3015, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27353310

RESUMEN

Schizophrenia (SZ) is a neurodevelopmental disorder in which the emergence of cognitive symptoms occurs during early adolescence. Glycogen synthase kinase-3ß (GSK3ß) plays a critical role in synaptic plasticity during development and is highly implicated in the etiology of SZ. However, how GSK3ß activity affects synaptic plasticity and working memory function in the prefrontal cortex (PFC) during development remains unknown. Here we show a GSK3ß hyperactivity during the early postnatal period in a neurodevelopmental rat SZ model that receives gestational exposure (E17) to the neurotoxin methylazoxymethanol (MAM). Accompanied with this change, adult MAM rats exhibited a significant decrease in spine density as well as impaired working memory, which was rescued by treatment with a GSK3ß inhibitor during the juvenile period. Furthermore, the age-dependent hyperactive GSK3ß caused a significant deficit in long-term potentiation (LTP) and facilitated long-term depression (LTD) in PFC pyramidal neurons. Notably, these changes in synaptic plasticity occurred only during the late juvenile period and were efficiently reversed by application of GSK3ß inhibitors. Because the balance of LTP and LTD plays a critical role in activity-dependent synaptic stabilization and elimination during cortical development, the transient hyperactive GSK3ß likely accounts for the cortical spine loss and PFC-dependent cognitive deficits in adulthood. These results highlight the importance of the postnatal trajectory of GSK3ß for spine development and PFC function, and may shed light on the prophylactic treatment of cognitive symptoms in the SZ.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta/metabolismo , Memoria a Corto Plazo/fisiología , Plasticidad Neuronal/fisiología , Neuronas/citología , Corteza Prefrontal/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Factores de Edad , Animales , Animales Recién Nacidos , Femenino , Técnicas In Vitro , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Acetato de Metilazoximetanol/farmacología , Plasticidad Neuronal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Embarazo , Inhibidores de la Síntesis de la Proteína/farmacología , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Eur Neuropsychopharmacol ; 25(10): 1723-32, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26271722

RESUMEN

It is now recognized that neurogenesis occurs throughout life predominantly in the subgranular zone (SGZ) of the hippocampus and the subventricular zone (SVZ) of the lateral ventricle. In the present study, we investigated the relationship between neurogenesis in the amygdala and extinction of fear memory. Mice received 15 tone-footshock pairings. Twenty-four hours after training, the mice were given 15 tone-alone trials (extinction training) once per day for 7 days. Two hours before extinction training, the mice were injected intraperitoneally with 5-bromo-3-deoxyuridine (BrdU). BrdU-positive and NeuN-positive cells were analyzed 52 days after the training. A group of mice that received tone-footshock pairings but no extinction training served as controls (FC+No-Ext). The number of BrdU(+)/NeuN(+) cells was significantly higher in the extinction (FC+Ext) than in the FC+No-Ext mice. Proliferation inhibitor methylazoxymethanol acetate (MAM) or DNA synthesis inhibitor cytosine arabinoside (Ara-C) reduced neurogenesis and retarded extinction. Silencing Sonic hedgehog (Shh) gene with short hairpin interfering RNA (shRNA) by means of a retrovirus expression system to knockdown Shh specifically in the mitotic neurons reduced neurogenesis and retarded extinction. By contrast, over-expression of Shh increased neurogenesis and facilitated extinction. These results suggest that amygdala neurogenesis and Shh signaling are involved in the extinction of fear memory.


Asunto(s)
Amígdala del Cerebelo/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Proteínas Hedgehog/metabolismo , Memoria/fisiología , Neurogénesis/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Antimitóticos/farmacología , Percepción Auditiva/efectos de los fármacos , Percepción Auditiva/fisiología , Citarabina/farmacología , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Proteínas Hedgehog/genética , Masculino , Memoria/efectos de los fármacos , Acetato de Metilazoximetanol/farmacología , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Distribución Aleatoria , Transducción de Señal/efectos de los fármacos
17.
Int J Neuropsychopharmacol ; 18(3)2014 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-25522410

RESUMEN

BACKGROUND: It is known that neurogenesis occurs throughout the life mostly in the subgranular zone of the hippocampus and the subventricular zone of the lateral ventricle. We investigated whether neurogenesis occurred in the amygdala and its function in fear memory formation. METHODS: For detection of newborn neurons, mice were injected intraperitoneally with 5-bromo-2'-deoxyuridine (BrdU) 2h before receiving 15 tone-footshock pairings, and newborn neurons were analyzed 14 and 42 days after training. To determine the relationship between neurogenesis and memory formation, mice were given a proliferation inhibitor methylazoxymethanol (MAM) or a DNA synthesis inhibitor cytosine arabinoside (Ara-C). To test whether sonic hedgehog (Shh) signaling was required for neurogenesis, Shh-small hairpin-interfering RNA (shRNA) was inserted into a retroviral vector (Retro-Shh-shRNA). RESULTS: The number of BrdU(+)/Neuronal nuclei (NeuN)(+) cells was significantly higher in the conditioned mice, suggesting that association of tone with footshock induced neurogenesis. MAM and Ara-C markedly reduced neurogenesis and impaired fear memory formation. Shh, its receptor patched 1 (Ptc1), and transcription factor Gli1 protein levels increased at 1 day and returned to baseline at 7 days after fear conditioning. Retro-Shh-shRNA, which knocked down Shh specifically in the mitotic neurons, reduced the number of BrdU(+)/NeuN(+) cells and decreased freezing responses. CONCLUSIONS: These results suggest that fear learning induces Shh signaling activation in the amygdala, which promotes neurogenesis and fear memory formation.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Condicionamiento Clásico/fisiología , Proteínas Hedgehog/metabolismo , Memoria a Largo Plazo/fisiología , Neurogénesis/fisiología , Transducción de Señal/fisiología , Animales , Bromodesoxiuridina , Citarabina/farmacología , Electrochoque , Conducta Exploratoria , Miedo/psicología , Inmunosupresores/farmacología , Potenciación a Largo Plazo , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño/farmacología
18.
J Neurosci ; 34(49): 16207-19, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25471562

RESUMEN

It has been recognized that the risk of cognitive decline during aging can be reduced if one maintains strong social connections, yet the neural events underlying this beneficial effect have not been rigorously studied. Here, we show that amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (APP/PS1) mice demonstrate improvement in memory after they are cohoused with wild-type mice. The improvement was associated with increased protein and mRNA levels of BDNF in the hippocampus. Concomitantly, the number of BrdU(+)/NeuN(+) cells in the hippocampal dentate gyrus was significantly elevated after cohousing. Methylazoxymethanol acetate, a cell proliferation blocker, markedly reduced BrdU(+) and BrdU/NeuN(+) cells and abolished the effect of social interaction. Selective ablation of mitotic neurons using diphtheria toxin (DT) and a retrovirus vector encoding DT receptor abolished the beneficial effect of cohousing. Knockdown of BDNF by shRNA transfection blocked, whereas overexpression of BDNF mimicked the memory-improving effect. A tropomyosin-related kinase B agonist, 7,8-dihydroxyflavone, occluded the effect of social interaction. These results demonstrate that increased BDNF expression and neurogenesis in the hippocampus after cohousing underlie the reversal of memory deficit in APP/PS1 mice.


Asunto(s)
Enfermedad de Alzheimer/terapia , Factor Neurotrófico Derivado del Encéfalo/fisiología , Hipocampo/metabolismo , Trastornos de la Memoria/terapia , Neurogénesis/fisiología , Conducta Social , Enfermedad de Alzheimer/complicaciones , Precursor de Proteína beta-Amiloide/genética , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Giro Dentado/fisiología , Toxina Diftérica/farmacología , Modelos Animales de Enfermedad , Flavonas/farmacología , Técnicas de Silenciamiento del Gen , Hipocampo/crecimiento & desarrollo , Vivienda para Animales , Masculino , Trastornos de la Memoria/complicaciones , Acetato de Metilazoximetanol/farmacología , Ratones , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Presenilina-1/genética
19.
Dev Neurobiol ; 74(10): 1002-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24723376

RESUMEN

Following development, the avian brain continues to produce neurons throughout adulthood, which functionally integrate throughout the telencephalon, including the hippocampus. In food-storing birds like the black-capped chickadee (Poecile atricapillus), new neurons incorporated into the hippocampus are hypothesized to play a role in spatial learning. Previous results on the relation between hippocampal neurogenesis and spatial learning, however, are correlational. In this study, we experimentally suppressed hippocampal neuronal recruitment and tested for subsequent effects on spatial learning in adult chickadees. After chickadees exhibited significant learning, we treated birds with daily injections of either saline or methylazoxymethanol (MAM), a toxin that suppresses cell proliferation in the brain and monitored subsequent spatial learning. MAM treatment significantly reduced cell proliferation around the lateral ventricles and neuronal recruitment in the hippocampus, measured using the cell birth marker bromodeoxyuridine. MAM-treated birds performed significantly worse than controls on the spatial learning task 12 days following the initiation of MAM treatment, a time when new neurons would begin functionally integrating into the hippocampus. This difference in learning, however, was limited to a single trial. MAM treatment did not affect any measure of body condition, suggesting learning impairments were not a product of non-specific adverse effects of MAM. This is the first evidence of a potential causal link between hippocampal neurogenesis and spatial learning in birds.


Asunto(s)
Hipocampo/fisiología , Neurogénesis/fisiología , Aprendizaje Espacial/fisiología , Animales , Bromodesoxiuridina/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Femenino , Hipocampo/efectos de los fármacos , Ventrículos Laterales , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/farmacología , Neurogénesis/efectos de los fármacos , Pruebas Neuropsicológicas , Distribución Aleatoria , Pájaros Cantores , Aprendizaje Espacial/efectos de los fármacos
20.
Eur Neuropsychopharmacol ; 24(2): 271-89, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23932495

RESUMEN

Several findings have indicated that schizophrenia may be connected with the impaired epigenetic regulation of gene transcription. The present study investigated the epigenetic modifications connected with histone H3 methylation at lysine (K)4 and K9 in the medial prefrontal cortex (mPFC) in a neurodevelopmental model of schizophrenia based on prenatal administration of methylazoxymethanol (MAM) at embryonic day 17, which impairs the sensorimotor gating process in adult but not adolescent animals. The effect of MAM was determined at different postnatal ages, pre-puberty (P15, P30 and P45) and post-puberty (P60 and P70), using western blot analyses. MAM treatment altered the levels of H3K9me2 before puberty. H3K9me2 was decreased at P15 and P45 but was increased at P30. In contrast, H3K4me3 was noticeably decreased in adult rats. Immunofluorescence experiments revealed that H3K9me2 protein levels were increased in neuronal cells at P30 and that H3K4me3 levels were decreased in astrocytes at P60 after MAM administration. Decreases in the methyltransferase ASH2L protein levels at P45, P60 and P70 were also observed, while the protein levels of the methyltransferase G9a did not change. In addition, levels of the demethylases LSD1 and JARID1c were analysed after MAM administration. LSD1 protein levels were increased at P15 but decreased at P30. JARID1c protein levels were increased in the MAM-treated animals at P60. Decreased Gad1 mRNA levels were found in adult MAM-treated animals, similar to alternation observed in schizophrenia. The present study indicates that prenatal MAM administration evokes changes in the methylation patterns of histone H3 during postnatal life.


Asunto(s)
Histonas/metabolismo , Acetato de Metilazoximetanol/análogos & derivados , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Teratógenos/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Peso Corporal/efectos de los fármacos , Encéfalo/anatomía & histología , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Inhibidores Enzimáticos/farmacología , Glutamato Descarboxilasa/metabolismo , Histona Demetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Acetato de Metilazoximetanol/farmacología , Metiltransferasas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Corteza Prefrontal/anatomía & histología , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Filtrado Sensorial/efectos de los fármacos , Ácido Valproico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...