Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-34445411

RESUMEN

BACKGROUND: The present study investigated the role of proteins from the bromodomain and extra-terminal (BET) family in schizophrenia-like abnormalities in a neurodevelopmental model of schizophrenia induced by prenatal methylazoxymethanol (MAM) administration (MAM-E17). METHODS: An inhibitor of BET proteins, JQ1, was administered during adolescence on postnatal days (P) 23-P29, and behavioural responses (sensorimotor gating, recognition memory) and prefrontal cortical (mPFC) function (long-term potentiation (LTP), molecular and proteomic analyses) studies were performed in adult males and females. RESULTS: Deficits in sensorimotor gating and recognition memory were observed only in MAM-treated males. However, adolescent JQ1 treatment affected animals of both sexes in the control but not MAM-treated groups and reduced behavioural responses in both sexes. An electrophysiological study showed LTP impairments only in male MAM-treated animals, and JQ1 did not affect LTP in the mPFC. In contrast, MAM did not affect activity-dependent gene expression, but JQ1 altered gene expression in both sexes. A proteomic study revealed alterations in MAM-treated groups mainly in males, while JQ1 affected both sexes. CONCLUSIONS: MAM-induced schizophrenia-like abnormalities were observed only in males, while adolescent JQ1 treatment affected memory recognition and altered the molecular and proteomic landscape in the mPFC of both sexes. Thus, transient adolescent inhibition of the BET family might prompt permanent alterations in the mPFC.


Asunto(s)
Azepinas/administración & dosificación , Acetato de Metilazoximetanol/análogos & derivados , Corteza Prefrontal/crecimiento & desarrollo , Esquizofrenia/fisiopatología , Triazoles/administración & dosificación , Adolescente , Desarrollo del Adolescente/efectos de los fármacos , Animales , Azepinas/farmacología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Acetato de Metilazoximetanol/toxicidad , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Proteómica , Ratas , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/inducido químicamente , Esquizofrenia/metabolismo , Caracteres Sexuales , Triazoles/farmacología
2.
Nicotine Tob Res ; 22(2): 204-212, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30899959

RESUMEN

INTRODUCTION: Patients with schizophrenia (SCZ) smoke at a rate of 4-5 times higher than the general population, contributing to negative health consequences in this group. One possible explanation for this increased smoking is that individuals with SCZ find nicotine (NIC) more reinforcing. However, data supporting this possibility are limited. METHODS: The present experiments examined self-administration of NIC, alone or in combination with other reinforcers, across a range of doses in the methylazoxymethanol acetate (MAM) rodent model of SCZ. RESULTS: MAM and control animals did not differ in NIC self-administration across a range of doses and schedules of reinforcement, in both standard 1-hour self-administration sessions and 23-hour extended access sessions. However, MAM animals responded less for sucrose or reinforcing visual stimuli alone or when paired with NIC. CONCLUSIONS: To the extent that MAM-treated rats are a valid model of SCZ, these results suggest that increased NIC reinforcement does not account for increased smoking in SCZ patients. IMPLICATIONS: This study is the first to utilize nicotine self-administration, the gold standard for studying nicotine reinforcement, in the methylazoxymethanol acetate model of schizophrenia, which is arguably the most comprehensive animal model of the disease currently available. Our assessment found no evidence of increased nicotine reinforcement in methylazoxymethanol acetate animals, suggesting that increased reinforcement may not perpetuate increased smoking in schizophrenia patients.


Asunto(s)
Modelos Animales de Enfermedad , Acetato de Metilazoximetanol/toxicidad , Nicotina/administración & dosificación , Refuerzo en Psicología , Esquizofrenia/inducido químicamente , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Inhibidores de la Síntesis de la Proteína/toxicidad , Ratas , Ratas Sprague-Dawley , Autoadministración , Sacarosa/administración & dosificación
3.
J Neurochem ; 143(3): 264-267, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28872674

RESUMEN

This Editorial highlights an article by Gulchina and colleagues in the current issue of the Journal of Neurochemistry, in which the authors describe molecular and epigenetic changes in the developing prefrontal cortex of the rats exposed to methylazoxymethanol acetate (MAM). They found an NMDAR hypofunction present in the prefrontal cortex of juvenile MAM rats which was associated with abnormal epigenetic regulation of the Grin2b gene. These changes may be related to early cognitive impairments observed in MAM rats and schizophrenia patients.


Asunto(s)
Epigénesis Genética/efectos de los fármacos , Acetato de Metilazoximetanol/toxicidad , Neurotoxinas/toxicidad , Corteza Prefrontal , Efectos Tardíos de la Exposición Prenatal , Esquizofrenia/etiología , Animales , Modelos Animales de Enfermedad , Femenino , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Esquizofrenia/patología
4.
J Neurochem ; 143(3): 320-333, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28628228

RESUMEN

Schizophrenia (SCZ) is characterized not only by psychosis, but also by working memory and executive functioning deficiencies, processes that rely on the prefrontal cortex (PFC). Because these cognitive impairments emerge prior to psychosis onset, we investigated synaptic function during development in the neurodevelopmental methylazoxymethanol (MAM) model for SCZ. Specifically, we hypothesize that N-methyl-D-aspartate receptor (NMDAR) hypofunction is attributable to reductions in the NR2B subunit through aberrant epigenetic regulation of gene expression, resulting in deficient synaptic physiology and PFC-dependent cognitive dysfunction, a hallmark of SCZ. Using western blot and whole-cell patch-clamp electrophysiology, we found that the levels of synaptic NR2B protein are significantly decreased in juvenile MAM animals, and the function of NMDARs is substantially compromised. Both NMDA-mEPSCs and synaptic NMDA-eEPSCs are significantly reduced in prelimbic PFC (plPFC). This protein loss during the juvenile period is correlated with an aberrant increase in enrichment of the epigenetic transcriptional repressor RE1-silencing transcription factor (REST) and the repressive histone marker H3K27me3 at the Grin2b promoter, as assayed by ChIP-quantitative polymerase chain reaction. Glutamate hypofunction has been a prominent hypothesis in the understanding of SCZ pathology; however, little attention has been given to the NMDAR system in the developing PFC in models for SCZ. Our work is the first to confirm that NMDAR hypofunction is a feature of early postnatal development, with epigenetic hyper-repression of the Grin2b promoter being a contributing factor. The selective loss of NR2B protein and subsequent synaptic dysfunction weakens plPFC function during development and may underlie early cognitive impairments in SCZ models and patients. Read the Editorial Highlight for this article on page 264.


Asunto(s)
Epigénesis Genética/fisiología , Corteza Prefrontal/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/patología , Animales , Animales Recién Nacidos , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Técnicas In Vitro , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/toxicidad , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Corteza Prefrontal/efectos de los fármacos , Embarazo , Ratas , Receptores de N-Metil-D-Aspartato/genética , Esquizofrenia/inducido químicamente , Esquizofrenia/complicaciones , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
5.
Arch Toxicol ; 91(10): 3427-3438, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28349193

RESUMEN

Genotoxic carcinogens pose great hazard to human health. Uncertainty of current risk assessment strategies and long latency periods between first carcinogen exposure and diagnosis of tumors have raised interest in predictive biomarkers. Initial DNA adduct formation is a necessary step for genotoxin induced carcinogenesis. However, as DNA adducts not always translate into tumorigenesis, their predictive value is limited. Here we hypothesize that the combined analysis of pro-mutagenic DNA adducts along with time-matched gene expression changes could serve as a superior prediction tool for genotoxic carcinogenesis. Eker rats, heterozygous for the tuberous sclerosis (Tsc2) tumor suppressor gene and thus highly susceptible towards genotoxic renal carcinogens, were continuously treated with the DNA alkylating carcinogen methylazoxymethanol acetate (MAMAc). Two weeks of MAMAc treatment resulted in a time-dependent increase of O6-methylguanine and N7-methylguanine adducts in the kidney cortex, which was however not reflected by significant expression changes of cyto-protective genes involved in DNA repair, cell cycle arrest or apoptosis. Instead, we found a transcriptional regulation of genes involved in the tumor-related MAPK, FoxO and TGF-beta pathways. Continuous MAMAc treatment for up to 6 months resulted in a mild but significant increase of cancerous lesions. In summary, the combined analysis of DNA adducts and early gene expression changes could serve as a suitable predictive tool for genotoxicant-induced carcinogenesis.


Asunto(s)
Aductos de ADN/análisis , Riñón/efectos de los fármacos , Acetato de Metilazoximetanol/toxicidad , Animales , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica , Guanina/análogos & derivados , Guanina/metabolismo , Riñón/metabolismo , Riñón/patología , Masculino , Acetato de Metilazoximetanol/administración & dosificación , Ratas Mutantes , Factores de Tiempo
6.
Cereb Cortex ; 24(4): 968-77, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23236209

RESUMEN

Schizophrenia is characterized by alterations in cortico-limbic processes believed to involve modifications in activity within the prefrontal cortex (PFC) and the hippocampus. The nucleus accumbens (NAc) integrates information from these 2 brain regions and is involved in cognitive and psychomotor functions that are disrupted in schizophrenia, indicating an important role for this structure in the pathophysiology of this disorder. In this study, we used in vivo electrophysiological recordings from the NAc and the PFC of adult rats and the MAM developmental disruption rodent model of schizophrenia to explore the influence of the medial PFC on the hippocampal-accumbens pathway. We found that, in MAM-treated rats, tetanization of hippocampal inputs to the NAc produce opposite synaptic plasticity compared with controls, which is a consequence of alterations in the hippocampal-mPFC pathway. Moreover, we show that administration of the D2-receptor-blocking antipsychotic drug sulpiride either systemically or directly into the mPFC reverses the alterations in the MAM rat. Therefore, specific disruptions in cortical and hippocampal inputs in the MAM-treated rat abnormally alter plasticity in subcortical structures. Moreover, our results suggest that, in the presence of antipsychotic drugs, the disrupted plasticities are normalized, supporting a role for this mechanism in antipsychotic drug action in schizophrenia.


Asunto(s)
Hipocampo/patología , Plasticidad Neuronal/fisiología , Núcleo Accumbens/patología , Corteza Prefrontal/patología , Esquizofrenia/patología , Sinapsis/patología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Acetato de Metilazoximetanol/toxicidad , Vías Nerviosas/patología , Neuronas/fisiología , Neurotoxinas/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Esquizofrenia/etiología
7.
Psychiatr Hung ; 30(1): 4-17, 2015.
Artículo en Húngaro | MEDLINE | ID: mdl-25867884

RESUMEN

Schizophrenia is a serious neuropsychiatric disorder. Several brain structures, neurotransmitter systems, genetic and environmental risk factors are suspected in the background. Because of its complexity the mechanism of the disorder is not known exactly, so the treatment of patients is unsolved. In the research of schizophrenia application of the rodent models is widespread. In this study one of these models based on the effect of methylazoxymethanol- acetate (MAM) is described, which is a neurodevelopmental, validated rat model. This antimitotic agent is able to evoke a number of schizophrenic symptomes temporarily disrupting the prenatal neurogenesis. The model reproduces numerous histological and neurophysiological changes of the human disorder, moreover it also represents several behavioral and cognitive phenomena resembling those in schizophrenia. A salient advantage of the model is the demonstration of the diachronic feature of the disorder, that is, postpubertal appearance of the positive symptoms. This model provides widespread opportunities for manipulations of the symptoms, so that using it in the future investigations can lead to a better understanding of this disorder.


Asunto(s)
Conducta Animal , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cognición , Modelos Animales de Enfermedad , Acetato de Metilazoximetanol/toxicidad , Neurotoxinas/toxicidad , Esquizofrenia/inducido químicamente , Esquizofrenia/fisiopatología , Psicología del Esquizofrénico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/fisiopatología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Sistema Límbico/metabolismo , Sistema Límbico/fisiopatología , Locomoción , Acetato de Metilazoximetanol/metabolismo , Trastornos del Movimiento/etiología , Trastornos del Movimiento/fisiopatología , Neurotoxinas/metabolismo , Ratas , Esquizofrenia/etiología , Esquizofrenia/metabolismo , Esquizofrenia/patología , Conducta Social
8.
J Neurosci ; 33(15): 6691-704, 2013 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-23575865

RESUMEN

Developmental perturbations during adolescence have been hypothesized to be a risk factor for the onset of several neuropsychiatric diseases. However the physiological alterations that result from such insults are incompletely understood. We investigated whether a defined perturbation during adolescence affected hippocampus-dependent sensorimotor gating functions, a proposed endophenotype in several psychiatric diseases, most notably schizophrenia. The developmental perturbation was induced during adolescence in mice using an antimitotic agent, methylazoxymethanol acetate (MAM), during postnatal weeks (PW) 4-6. MAM-treated mice showed a decrease in hippocampal neurogenesis immediately after treatment, which was restored by PW10 in adulthood. However, the mice treated with MAM during adolescent stages exhibited a persistent sensorimotor gating deficiency and a reduction in prepulse inhibition-related activation of hippocampal and prefrontal neurons in adulthood. Cellular analyses found a reduction of GABAergic inhibitory neurons and abnormal dendritic morphology of immature neurons in the dentate gyrus (DG). Interestingly, bilateral infusion of muscimol, a GABAA receptor agonist, into the DG region reversed the prepulse inhibition abnormality in MAM-treated mice. Furthermore, the behavioral deficits together with the decrease in the number of GABAergic neurons in this MAM model were rescued by exposure to an enriched environment during a defined critical adolescent period. These observations suggest a possible role for GABAergic interneurons in the DG during adolescence. This role may be related to the establishment of neural circuitry required for sensorimotor gating. It is plausible that changes in neurogenesis during this window may affect the survival of GABAergic interneurons, although this link needs to be causally addressed.


Asunto(s)
Período Crítico Psicológico , Giro Dentado/fisiología , Neuronas GABAérgicas/fisiología , Interneuronas/fisiología , Acetato de Metilazoximetanol/toxicidad , Filtrado Sensorial/fisiología , Factores de Edad , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Dendritas , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Quinasas Similares a Doblecortina , Interacciones Farmacológicas , Ambiente , Agonistas del GABA/administración & dosificación , Agonistas del GABA/farmacología , Neuronas GABAérgicas/metabolismo , Inhibición Psicológica , Interneuronas/citología , Interneuronas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Proteínas Asociadas a Microtúbulos/metabolismo , Muscimol/administración & dosificación , Muscimol/farmacología , Muscimol/uso terapéutico , Neurogénesis/efectos de los fármacos , Neuropéptidos/metabolismo , Corteza Prefrontal/fisiología , Proteínas Serina-Treonina Quinasas , Esquizofrenia/inducido químicamente , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología
9.
J Neurophysiol ; 111(2): 323-35, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24155012

RESUMEN

Cortical dysplasia (CD) associates with clinical pathologies, including epilepsy and mental retardation. CD results from impaired migration of immature neurons to their cortical targets, leading to clustering of neural cells and changes in cortical properties. We developed a CD model by administering methylazoxymethanol (MAM), an anti-mitotic, to pregnant ferrets on embryonic day 33; this leads to reduction in cortical thickness in addition to redistribution and increased expression of GABAA receptors (GABAAR). We evaluated the impact of MAM treatment on GABAAR-mediated synaptic transmission in postnatal day 0-1 neurons, leaving the ganglionic eminence (GE) and in layer 2/3 pyramidal cells of postnatal day 28-38 ferrets. Embryonic day 33 MAM treatment significantly increases the amplitude and frequency of spontaneous GABAAR-mediated inhibitory postsynaptic currents (IPSCs) in the cells leaving the GE. In older MAM-treated animals, the amplitude and frequency of GABAAR-mediated spontaneous IPSCs in layer 2/3 pyramidal cells is increased, as are the amplitude and frequency of miniature IPSCs. The kinetics of GABAAR opening also altered following treatment with MAM. Western blot analysis shows that the expression of the GABAAα3R and GABAAγ2R subunits amplified in our model animals. We did not observe any significant change in the passive properties of either the layer 2/3 pyramidal cells or cells leaving the GE after MAM treatment. These observations reinforce the idea that synaptic neurotransmission through GABAAR enhances following treatment with MAM and coincides with our finding of increased GABAAαR expression within the upper cortical layers. Overall, we demonstrate that small amounts of toxins delivered during corticogenesis can result in long-lasting changes in ambient expression of GABAAR that influence intrinsic neuronal properties.


Asunto(s)
Potenciales Postsinápticos Inhibidores , Malformaciones del Desarrollo Cortical/fisiopatología , Potenciales Postsinápticos Miniatura , Neocórtex/fisiopatología , Receptores de GABA-A/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hurones , Malformaciones del Desarrollo Cortical/inducido químicamente , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/toxicidad , Neocórtex/citología , Neocórtex/embriología , Neocórtex/metabolismo , Embarazo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células Piramidales/metabolismo , Células Piramidales/fisiología , Receptores de GABA-A/genética , Teratógenos/toxicidad
10.
Int J Neuropsychopharmacol ; 17(10): 1609-19, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24787547

RESUMEN

Alteration in normal hippocampal (HPC) function attributed to reduced parvalbumin (PV) expression has been consistently reported in schizophrenia patients and in animal models of schizophrenia. However, it is unclear whether there is an overall loss of interneurons as opposed to a reduction in activity-dependent PV content. Co-expression of PV and the constitutively expressed substance P (SP)-receptor protein has been utilized in other models to ascertain the degree of cell survival, as opposed to reduction in activity-dependent PV content, in the HPC. The present study measured the co-expression of PV and SP-receptors in the dentate and dorsal and ventral CA3 subregions of the HPC in the methylazoymethanol acetate (MAM) rat neurodevelopmental model of schizophrenia. In addition, these changes were compared at the post-natal day 27 (PND27) and post-natal day 240 (PND > 240) time points. Brains from PND27 and PND > 240 MAM (n = 8) and saline (SAL, n = 8) treated offspring were immunohistochemically processed for the co-expression of PV and SP-receptors. The dorsal dentate, dorsal CA3 and ventral CA3 subregions of PND27 and PND > 240 MAM rats demonstrated significant reductions in PV but not SP-receptor expression, signifying a loss of PV-content. In contrast, in the ventral dentate the co-expression of PV and SP-receptors was significantly reduced only in PND > 240 MAM animals, suggesting a reduction in cell number. While MAM-induced reduction of PV content occurs in CA3 of dorsal and ventral HPC, the most substantial loss of interneuron number is localized to the ventral dentate of PND > 240 animals. The disparate loss of PV in HPC subregions likely impacts intra-HPC network activity in MAM rats.


Asunto(s)
Hipocampo/patología , Neuronas/metabolismo , Parvalbúminas/metabolismo , Receptores de Neuroquinina-1/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/patología , Factores de Edad , Análisis de Varianza , Animales , Muerte Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Acetato de Metilazoximetanol/toxicidad , Neuronas/efectos de los fármacos , Neuronas/patología , Neurotoxinas/toxicidad , Ratas , Esquizofrenia/inducido químicamente
11.
Arch Toxicol ; 88(1): 127-36, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23917397

RESUMEN

Developmental neurotoxicity (DNT) of environmental chemicals is a serious threat to human health. Current DNT testing guidelines propose investigations in rodents, which require large numbers of animals. With regard to the "3Rs" (reduction, replacement, and refinement) of animal testing, alternative testing strategies are needed in order to refine and reduce animal experiments and allow faster and less expensive screening. The goal of this study was to establish components for a human cell-based test system to assess DNT potential of chemicals at an early stage of brain development. A human neural precursor cell line should be tested for suitability for semi-automated high-throughput DNT screening. We established assays suitable for detecting disturbances in two basic processes of brain development in 96-well scale: neuronal differentiation and migration using the human Ntera2 (NT2) cell line. We assessed the effects of four test compounds with well-established DNT potential in comparison with three compounds without specific DNT potential. We found that human NT2 cell cultures treated with the morphogen, retinoic acid, imitate neuronal differentiation, and migration in vitro. The developmental neurotoxicants methylmercury chloride, sodium arsenite, sodium valproate, and methylazoxymethanol significantly reduced the expression of the neuronal marker ß-tubulin type III and decreased the migration distance in developing NT2 cells. Both endpoints, differentiation and migration, can be read out directly in a standard fluorescence plate reader, enabling high-throughput screening. We conclude that NT2 cell tests are likely to become valuable components of a human cell-based modular in vitro DNT test systems.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Ensayos Analíticos de Alto Rendimiento/métodos , Síndromes de Neurotoxicidad/etiología , Pruebas de Toxicidad/métodos , Arsenitos/toxicidad , Encéfalo/efectos de los fármacos , Diferenciación Celular , Línea Celular , Movimiento Celular , Humanos , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/toxicidad , Compuestos de Metilmercurio/toxicidad , Neuronas/citología , Neuronas/efectos de los fármacos , Compuestos de Sodio/toxicidad , Tubulina (Proteína)/metabolismo , Ácido Valproico/toxicidad
12.
Eur Neuropsychopharmacol ; 78: 30-42, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37866191

RESUMEN

Social and cognitive dysfunctions are the most persistent symptoms of schizophrenia. Since oxytocin (OXT) is known to play a role in social functions and modulates cognitive processes, we investigated the effects of a novel, nonpeptide, selective OXT receptor agonist, LIT-001, in a neurodevelopmental model of schizophrenia. Administration of methylazoxymethanol acetate (MAM; 22 mg/kg) on the 17th day of rat pregnancy is known to cause developmental disturbances of the brain, which lead to schizophrenia-like symptomatology in the offspring. Here, we examined the effects of acutely administered LIT-001 (1, 3, and 10 mg/kg) in MAM-exposed males and females on social behaviour, communication and cognition. We report that MAM-treated adult male and female rats displayed reduced social behaviour, ultrasonic communication and novel object recognition test performance. LIT-001 partially reversed these deficits, increasing the total social interaction time and the number of 'positive', highly-modulated 50 kHz ultrasonic calls in male rats. The compound ameliorated MAM-induced deficits in object discrimination in both sexes. Present results confirm the pro-social activity of LIT-001 and demonstrate its pro-cognitive effects following acute administration.


Asunto(s)
Pirazoles , Pirrolidinas , Esquizofrenia , Embarazo , Ratas , Femenino , Masculino , Animales , Esquizofrenia/inducido químicamente , Esquizofrenia/tratamiento farmacológico , Oxitocina/farmacología , Receptores de Oxitocina , Cognición , Acetato de Metilazoximetanol/toxicidad , Modelos Animales de Enfermedad
13.
CNS Neurosci Ther ; 30(2): e14565, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38421095

RESUMEN

AIM: Widely used second-generation antipsychotics are associated with adverse metabolic effects, contributing to increased cardiovascular mortality. To develop strategies to prevent or treat adverse metabolic effects, preclinical models have a clear role in uncovering underlying molecular mechanisms. However, with few exceptions, preclinical studies have been performed in healthy animals, neglecting the contribution of dysmetabolic features inherent to psychotic disorders. METHODS: In this study, methylazoxymethanol acetate (MAM) was prenatally administered to pregnant Sprague-Dawley rats at gestational day 17 to induce a well-validated neurodevelopmental model of schizophrenia mimicking its assumed pathogenesis with persistent phenotype. Against this background, the dysmetabolic effects of acute treatment with olanzapine and haloperidol were examined in female rats. RESULTS: Prenatally MAM-exposed animals exhibited several metabolic features, including lipid disturbances. Half of the MAM rats exposed to olanzapine had pronounced serum lipid profile alteration compared to non-MAM controls, interpreted as a reflection of a delicate MAM-induced metabolic balance disrupted by olanzapine. In accordance with the drugs' clinical metabolic profiles, olanzapine-associated dysmetabolic effects were more pronounced than haloperidol-associated dysmetabolic effects in non-MAM rats and rats exposed to MAM. CONCLUSION: Our results demonstrate metabolic vulnerability in female prenatally MAM-exposed rats, indicating that findings from healthy animals likely provide an underestimated impression of metabolic dysfunction associated with antipsychotics. In the context of metabolic disturbances, neurodevelopmental models possess a relevant background, and the search for adequate animal models should receive more attention within the field of experimental psychopharmacology.


Asunto(s)
Antipsicóticos , Haloperidol , Acetato de Metilazoximetanol/análogos & derivados , Embarazo , Ratas , Femenino , Animales , Haloperidol/toxicidad , Acetato de Metilazoximetanol/toxicidad , Olanzapina/toxicidad , Ratas Sprague-Dawley , Antipsicóticos/uso terapéutico , Lípidos , Modelos Animales de Enfermedad
14.
Exp Neurol ; 376: 114759, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38519010

RESUMEN

Malformations of cortical development (MCDs) are caused by abnormal neuronal migration processes during the fetal period and are a major cause of intractable epilepsy in infancy. However, the timing of hyperexcitability or epileptogenesis in MCDs remains unclear. To identify the early developmental changes in the brain of the MCD rat model, which exhibits increased seizure susceptibility during infancy (P12-15), we analyzed the pathological changes in the brains of MCD model rats during the neonatal period and tested NMDA-induced seizure susceptibility. Pregnant rats were injected with two doses of methylazoxymethanol acetate (MAM, 15 mg/kg, i.p.) to induce MCD, while controls were administered normal saline. The cortical development of the offspring was measured by performing magnetic resonance imaging (MRI) on postnatal days (P) 1, 5, and 8. At P8, some rats were sacrificed for immunofluorescence, Golgi staining, and Western analysis. In another set of rats, the number and latency to onset of spasms were monitored for 90 min after the NMDA (5 mg/kg i.p.) injection at P8. In MCD rats, in vivo MR imaging showed smaller brain volume and thinner cortex from day 1 after birth (p < 0.001). Golgi staining and immunofluorescence revealed abnormal neuronal migration, with a reduced number of neuronal cell populations and less dendritic arborization at P8. Furthermore, MCD rats exhibited a significant reduction in the expression of NMDA receptors and AMPAR4, along with an increase in AMPAR3 expression (p < 0.05). Although there was no difference in the latency to seizure onset between MCD rats and controls, the MCD rats survived significantly longer than the controls. These results provide insights into the early developmental changes in the cortex of a MCD rat model and suggest that delayed and abnormal neuronal development in the immature brain is associated with a blunted response to NMDA-induced excitotoxic injury. These developmental changes may be involved in the sudden onset of epilepsy in patients with MCD or prenatal brain injury.


Asunto(s)
Movimiento Celular , Modelos Animales de Enfermedad , Malformaciones del Desarrollo Cortical , N-Metilaspartato , Neuronas , Ratas Sprague-Dawley , Animales , Ratas , N-Metilaspartato/toxicidad , Femenino , Embarazo , Movimiento Celular/efectos de los fármacos , Neuronas/patología , Neuronas/efectos de los fármacos , Malformaciones del Desarrollo Cortical/inducido químicamente , Malformaciones del Desarrollo Cortical/patología , Animales Recién Nacidos , Acetato de Metilazoximetanol/toxicidad , Acetato de Metilazoximetanol/análogos & derivados , Corteza Cerebral/patología , Corteza Cerebral/efectos de los fármacos , Masculino , Imagen por Resonancia Magnética
15.
Int J Neuropsychopharmacol ; 16(6): 1331-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23190686

RESUMEN

Subcortical dopamine system dysregulation has been suggested to underlie the positive symptoms of schizophrenia. Recent preclinical investigations and human imaging studies have proposed that the augmented dopamine system function observed in schizophrenia patients may be secondary to aberrant hippocampal activity. Thus, we posit that the hippocampus represents a novel therapeutic target for the treatment of schizophrenia. Here we provide evidence of the effectiveness of a unique approach aimed at decreasing hippocampal function in a rodent model of schizophrenia. Specifically, in a rodent model of schizophrenia, we demonstrate that ventral hippocampal (vHipp) deep brain stimulation (DBS) can normalize aberrant dopamine neuron activity and behaviours associated with positive symptoms. In addition, we provide evidence that this approach may also be effective in restoring deficits in cognitive function, often left unaltered by conventional antipsychotic medications. Therefore, we have provided initial preclinical evidence demonstrating the feasibility of hippocampal DBS as a potential novel approach for the treatment of schizophrenia.


Asunto(s)
Estimulación Encefálica Profunda/métodos , Hipocampo/fisiología , Trastornos Mentales/terapia , Esquizofrenia/complicaciones , Potenciales de Acción/efectos de los fármacos , Anfetamina/efectos adversos , Animales , Animales Recién Nacidos , Trastorno por Déficit de Atención con Hiperactividad/etiología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/fisiología , Femenino , Hipercinesia/inducido químicamente , Masculino , Trastornos Mentales/etiología , Acetato de Metilazoximetanol/toxicidad , Neurotoxinas/toxicidad , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Esquizofrenia/etiología , Esquizofrenia/terapia , Área Tegmental Ventral/patología
16.
Int J Neuropsychopharmacol ; 16(3): 507-12, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23067577

RESUMEN

The nucleus accumbens (NAc) receives converging inputs from the medial prefrontal cortex (mPFC) and the hippocampus which have competitive interactions in the NAc to influence motivational drive. We have previously shown altered synaptic plasticity in the hippocampal-NAc pathway in the methylazoxymethanol acetate (MAM) developmental model of schizophrenia in rodents that is dependent on cortical inputs. Thus, because mPFC-hippocampal balance is known to be partially altered in this model, we investigated potential pathological changes in the hippocampal influence over cortex-driven NAc spike activity. Here we show that the reciprocal interaction between the hippocampus and mPFC is absent in MAM animals but is able to be reinstated with administration of the antipsychotic drug, sulpiride. The lack of interaction between these structures in this model could explain the attentional deficits in schizophrenia patients and shed light onto their inability to focus on a single task.


Asunto(s)
Antipsicóticos/uso terapéutico , Modelos Animales de Enfermedad , Hipocampo/fisiología , Corteza Prefrontal/fisiología , Esquizofrenia/tratamiento farmacológico , Sulpirida/uso terapéutico , Animales , Antipsicóticos/farmacología , Femenino , Hipocampo/efectos de los fármacos , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Acetato de Metilazoximetanol/toxicidad , Corteza Prefrontal/efectos de los fármacos , Embarazo , Ratas , Ratas Sprague-Dawley , Esquizofrenia/inducido químicamente , Esquizofrenia/fisiopatología , Sulpirida/farmacología
17.
Schizophr Res ; 262: 32-39, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37922841

RESUMEN

Schizophrenia is a neurodevelopmental psychiatric disorder that often emerges in adolescence, is characterized by social dysfunction, and has an earlier onset in men. These features have been replicated in rats exposed to the mitotoxin methylazoxymethanol acetate (MAM) on gestational day (GD) 17, which as adults exhibit behavioral impairments and dopamine (DA) system changes consistent with a schizophrenia-relevant rodent model. In humans, social withdrawal is a negative symptom that often precedes disease onset and DA system dysfunction and is more pronounced in men. Children and adolescents at high-risk for schizophrenia exhibit social deficits prior to psychotic symptoms (i.e., prodromal phase), which can be used as a predictive marker for future psychopathology. Adult MAM rats also exhibit deficient social interaction, but less is known regarding the emergence of social dysfunction in this model, whether it varies by sex, and whether it is linked to disrupted DA function. To this end, we characterized the ontogeny of social and DA dysfunction in male and female MAM rats during the prepubertal period (postnatal days 33-43) and found sex-specific changes in motivated social behaviors (play, approach) and DA function. Male MAM rats exhibited reduced social approach and increased VTA DA neuron activity compared to saline-treated (SAL) males, whereas female MAM rats exhibited enhanced play behaviors compared to SAL females but no changes in social approach or VTA population activity during this period. These findings demonstrate sex differences in the emergence of social and DA deficits in the MAM model, in which females exhibit delayed emergence.


Asunto(s)
Dopamina , Esquizofrenia , Humanos , Adolescente , Niño , Ratas , Masculino , Femenino , Animales , Dopamina/fisiología , Esquizofrenia/inducido químicamente , Roedores , Acetato de Metilazoximetanol/toxicidad , Neuronas , Modelos Animales de Enfermedad
18.
J Neurosci ; 31(34): 12330-8, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21865475

RESUMEN

Repeated administration of antipsychotic drugs to normal rats has been shown to induce a state of dopamine neuron inactivation known as depolarization block, which correlates with the ability of the drugs to exhibit antipsychotic efficacy and extrapyramidal side effects in schizophrenia patients. Nonetheless, in normal rats depolarization block requires weeks of antipsychotic drug administration, whereas schizophrenia patients exhibit initial effects soon after initiating antipsychotic drug treatment. We now report that, in a developmental disruption rat model of schizophrenia [methyl-azoxymethanol acetate (20 mg/kg, i.p.) injected into G17 pregnant female rats, with offspring tested as adults], the extant hyperdopaminergic state combines with the excitatory actions of a first- (haloperidol; 0.6 mg/kg, i.p.) and a second- (sertindole; 2.5 mg/kg, i.p.) generation antipsychotic drug to rapidly induce depolarization block in ventral tegmental area dopamine neurons. Acute injection of either antipsychotic drug induced an immediate reduction in the number of spontaneously active dopamine neurons (cells per electrode track; termed population activity). Repeated administration of either antipsychotic drug for 1, 3, 7, 15, and 21 d continued to reduce dopamine neuron population activity. Both acute and repeated effects on population activity were reversed by acute apomorphine injections, which is consistent with the reversal of dopamine neuron depolarization block. Although this action may account for the effects of D2 antagonist drugs on alleviating psychosis and the lack of development of tolerance in humans, the drugs appear to do so by inducing an offsetting deficit rather than attacking the primary pathology present in schizophrenia.


Asunto(s)
Antipsicóticos/farmacología , Dopamina/fisiología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Área Tegmental Ventral/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Polaridad Celular/efectos de los fármacos , Polaridad Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Acetato de Metilazoximetanol/toxicidad , Inhibición Neural/fisiología , Neuronas/metabolismo , Neurotoxinas/toxicidad , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología
19.
Synapse ; 65(5): 393-403, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20803618

RESUMEN

Schizophrenia is a highly familial, neurodevelopmental disorder that is associated with several neuropsychiatric, psychological, and neuropathological features. Although pharmacological animal models of dopaminergic and glutamatergic dysfunction have helped advance our understanding of the disease biology, there is a clear need for translational models that capture the neuropathological and functional manifestations associated with the intermediate phenotype and the clinical illness. Neuroimaging of preclinical neurodevelopmental approaches such as methylazoxymethanol acetate (MAM) exposure may afford a powerful translational tool to establish endpoints with greater congruency across animals and humans. Using in vivo volumetric magnetic resonance imaging (MRI), manganese-enhanced MRI, and diffusion tensor imaging (DTI), we investigated morphological and cytoarchitectural changes of brain structures in MAM-exposed rats, a neurodevelopmental model of schizophrenia. Compared to saline-exposed controls, MAM-exposed rats showed significant enlargement of lateral and third ventricles as well as reduced hippocampal volumes, which is consistent with findings observed in schizophrenia. In addition, DTI revealed that diffusion fractional anisotropy retrieved from corpus callosum and cingulum were significantly decreased in MAM-exposed rats, suggesting that demyelination occurred in these white-matter fiber tracts. Imaging findings were confirmed by conducting histological analysis using hematoxylin and eosin and Luxol fast blue stainings. In summary, structural abnormalities resulting from a MAM environmental challenge parallel cerebral pathology observed in schizophrenia. The MAM model incorporating noninvasive imaging techniques may therefore serve as an improved translational research tool for assessing new treatments for schizophrenia.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Acetato de Metilazoximetanol/toxicidad , Neurotoxinas/toxicidad , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Factores de Edad , Animales , Animales Recién Nacidos , Mapeo Encefálico , Corteza Cerebral/crecimiento & desarrollo , Ventrículos Cerebrales/crecimiento & desarrollo , Ventrículos Cerebrales/patología , Cuerpo Calloso/crecimiento & desarrollo , Cuerpo Calloso/patología , Imagen de Difusión por Resonancia Magnética , Femenino , Hipocampo/crecimiento & desarrollo , Hipocampo/patología , Imagen por Resonancia Magnética/métodos , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley
20.
Epilepsia ; 52(12): 2304-14, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21933177

RESUMEN

PURPOSE: Periventricular nodular heterotopia (PNH) is, in humans, often associated with difficult-to-control epilepsy. However, there is considerable controversy about the role of the PNH in seizure generation and spread. To study this issue, we have used a rat model in which injection of methylazoxymethanol (MAM) into pregnant rat dams produces offspring with nodular heterotopia-like brain abnormalities. METHODS: Electrophysiologic methods were used to examine the activity of the MAM-induced PNH relative to activity in the neighboring hippocampus and overlying neocortex. Recordings were obtained simultaneously from these three structures in slice preparations from MAM-exposed rats and in intact animals. Bath application or systemic injection of bicuculline was used to induce epileptiform activity. KEY FINDINGS: In the in vitro slice, epileptiform discharge was generally initiated in hippocampus. In some cases, independent PNH discharge occurred, but the PNH never "led" discharges in hippocampus or neocortex. Intracellular recordings from PNH neurons confirmed that these cells received synaptic drive from both hippocampus and neocortex, and sent axonal projections to these structures-consistent with anatomic observations of biocytin-injected PNH cells. In intact animal preparations, bicuculline injection resulted in epileptiform discharge in all experiments, with a period of ictal-like electrographic activity typically initiated within 2-3 min after drug injection. In almost all animals, the onset of ictus was seen synchronously across PNH, hippocampal, and neocortical electrodes; in a few cases, the PNH electrode (histologically confirmed) did not participate, but in no case was activity initiated in the PNH electrode. Interictal discharge was also synchronized across all three electrodes; again, the PNH never "led" the other two electrodes, and typically followed (onset several milliseconds after hippocampal/neocortical discharge onset). SIGNIFICANCE: These results do not support the hypothesis that the PNH lesion is the primary epileptogenic site, since it does not initiate or lead epileptiform activity that subsequently propagates to other brain regions.


Asunto(s)
Modelos Animales de Enfermedad , Epilepsia/etiología , Heterotopia Nodular Periventricular/complicaciones , Potenciales de Acción/efectos de los fármacos , Animales , Femenino , Hipocampo/patología , Hipocampo/fisiopatología , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/efectos de los fármacos , Masculino , Acetato de Metilazoximetanol/análogos & derivados , Acetato de Metilazoximetanol/toxicidad , Neocórtex/patología , Neocórtex/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Heterotopia Nodular Periventricular/inducido químicamente , Heterotopia Nodular Periventricular/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Teratógenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA