Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cells ; 10(12)2021 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-34944011

RESUMEN

Both in utero exposure to maternal immune activation and cannabis use during adolescence have been associated with increased risk for the development of schizophrenia; however, whether these exposures exert synergistic effects on brain function is not known. In the present study, mild maternal immune activation (MIA) was elicited in mice with prenatal exposure to polyinosinic-polycytidylic acid (poly(I:C)), and ∆9-tetrahydrocannabinol (THC) was provided throughout adolescence in cereal (3 mg/kg/day for 5 days). Neither THC nor MIA pretreatments altered activity in assays used to characterize hyperdopaminergic states in adulthood: amphetamine hyperlocomotion and prepulse inhibition of the acoustic startle reflex. Adolescent THC treatment elicited deficits in spatial memory and enhanced spatial reversal learning in adult female mice in the Morris water maze, while exposure to MIA elicited female-specific deficits in fear extinction learning in adulthood. There were no effects in these assays in adult males, nor were there interactions between THC and MIA in adult females. While doses of poly(I:C) and THC were sufficient to elicit behavioral effects, particularly relating to cognitive performance in females, there was no evidence that adolescent THC exposure synergized with the risk imposed by MIA to worsen behavioral outcomes in adult mice of either sex.


Asunto(s)
Envejecimiento/fisiología , Conducta Animal/efectos de los fármacos , Dronabinol/farmacología , Efectos Tardíos de la Exposición Prenatal/inmunología , Anfetamina , Animales , Condicionamiento Clásico , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Femenino , Locomoción/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Embarazo , Inhibición Prepulso/efectos de los fármacos , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Natación
2.
Int J Neuropsychopharmacol ; 24(11): 894-906, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34338765

RESUMEN

BACKGROUND: HIV-associated neurocognitive disorder (HAND) is commonly observed in persons living with HIV (PWH) and is characterized by cognitive deficits implicating disruptions of fronto-striatal neurocircuitry. Such circuitry is also susceptible to alteration by cannabis and other drugs of abuse. PWH use cannabis at much higher rates than the general population, thus prioritizing the characterization of any interactions between HIV and cannabinoids on cognitively relevant systems. Prepulse inhibition (PPI) of the startle response, the process by which the motor response to a startling stimulus is attenuated by perception of a preceding non-startling stimulus, is an operational assay of fronto-striatal circuit integrity that is translatable across species. PPI is reduced in PWH. The HIV transgenic (HIVtg) rat model of HIV infection mimics numerous aspects of HAND, although to date the PPI deficit observed in PWH has yet to be fully recreated in animals. METHODS: PPI was measured in male and female HIVtg rats and wild-type controls following acute, nonconcurrent treatment with the primary constituents of cannabis: Δ 9-tetrahydrocannabinol (THC; 1 and 3 mg/kg, s.c.) and cannabidiol (1, 10, and 30 mg/kg, i.p.). RESULTS: HIVtg rats exhibited a significant PPI deficit relative to wild-type controls. THC reduced PPI in controls but not HIVtg rats. Cannabidiol exerted only minor, genotype-independent effects on PPI. CONCLUSIONS: HIVtg rats exhibit a relative insensitivity to the deleterious effects of THC on the fronto-striatal function reflected by PPI, which may partially explain the higher rates of cannabis use among PWH.


Asunto(s)
Cannabinoides/farmacología , Infecciones por VIH/fisiopatología , Filtrado Sensorial/efectos de los fármacos , Estimulación Acústica , Animales , Cannabidiol/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Dronabinol/farmacología , Femenino , Alucinógenos/farmacología , Masculino , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Transgénicas , Reflejo de Sobresalto/efectos de los fármacos
3.
Neurosci Lett ; 755: 135913, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33895274

RESUMEN

Schizophrenia modeling by disrupting prepulse inhibition (PPI) is one of the most frequently used psycho-pharmacological methods by administering pharmacological agents to stimulate disruption. However, since PPI is also a biological indicator of schizophrenia, it is possible to classify subjects based on their basal PPI values and group them as "low inhibition" and "high inhibition without taking any pharmacological agent. Therefore this study was conducted to show that rats can be divided into groups in terms of susceptibility to schizophrenia according to basal PPI values. It was also observed that these groups might give different responses to different pharmacological agents (apomorphine, amphetamine, MK-801, scopolamine, nicotine, caffeine). Male Sprague Dawley rats (250-350 g) were used in the study. To examine the effects of different pharmacological agents on the groups, apomorphine (0.5 mg/kg and 1 mg/kg), amphetamine (4 mg/kg), MK-801 (0.05 mg/kg and 0.15 mg/kg), scopolamine (0.4 mg/kg), nicotine (1 mg/kg) and caffeine (10 mg/kg and 30 mg/kg) were used. Amphetamine showed a disruptive effect on PPI in both low and high inhibitory groups, while apomorphine, MK-801, scopolamine, and nicotine showed PPI decrease only in the high inhibitory group. Besides, caffeine decreased PPI levels at two doses in the high inhibitory group; however, 10 mg/kg dose caffeine was increased only in the low inhibitory group. According to the data obtained from this study, rats can be grouped with baseline inhibition values by using PPI, and response differences of pharmacological agents to groups may vary.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Antagonistas Colinérgicos/farmacología , Agonistas de Dopamina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Agonistas Nicotínicos/farmacología , Inhibición Prepulso/efectos de los fármacos , Estimulación Acústica/métodos , Anfetamina/farmacología , Animales , Apomorfina/farmacología , Cafeína/farmacología , Maleato de Dizocilpina/farmacología , Masculino , Nicotina/farmacología , Inhibición Prepulso/fisiología , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Escopolamina/farmacología
4.
Behav Neurosci ; 135(1): 32-38, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33734732

RESUMEN

Prepulse inhibition (PPI) refers to the modulation of the startle response by the presentation of a weaker stimulus prior to the onset of the startle stimulus. This response is consolidated along the maturation process of the mesocortical system, where the dopamine neurotransmitter plays an important role. In fact, it has been reported that agonist and antagonist dopaminergic drugs are able to change PPI expression. This study was aimed to analyze the relationship between the adult medial prefrontal cortex (mPfc) and dopaminergic involvement in PPI throughout the life span. Specifically, the present experiment analyzed the effect of the administration of dopaminergic agonist amphetamine on PPI in two different age periods in Wistar rats: postnatal day (PND) 28 and PND 70. In this last period, we also explored the relationship between PPI response and amphetamine effects after mPfc lesion. The results showed that PPI was expressed in all groups and periods; however, amphetamine only modulated this effect during adulthood. We also found that the mPfc is essential to modulate PPI after amphetamine consumption. Besides, our results suggest a role for dopamine and mPfc as important modulators of PPI in adulthood. Nevertheless, this neurotransmitter could not be involved in the expression of PPI because the administration of a dopaminergic agonist was ineffective in PND-28 period. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Asunto(s)
Anfetamina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Inhibición Prepulso/efectos de los fármacos , Estimulación Acústica , Animales , Dopamina/metabolismo , Dopaminérgicos/farmacología , Masculino , Ratas , Ratas Wistar , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología
5.
Neurotox Res ; 39(3): 543-555, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33608816

RESUMEN

The effects of permethrin (PRM) and deltamethrin (DLM) on acoustic or light prepulse inhibition of the acoustic startle response (ASR) and tactile startle response (TSR) were studied in adult male Sprague Dawley rats. Preliminary studies were conducted to optimize the parameters of light and acoustic prepulse inhibition of ASR and TSR. Once these parameters were set, a new group of rats was administered PRM (0 or 90 mg/kg) or DLM (0 or 25 mg/kg) by gavage in 5 mL/kg corn oil. ASR and TSR were assessed using acoustic or light prepulses 6, 8, and 12 h after PRM and 2, 4, and 6 h after DLM exposure. PRM increased ASR 6 h post-treatment with no interaction with acoustic prepulse levels and with no effect on TSR. When light was used as the prepulse, PRM increased ASR and TSR at 6 h with no interaction with prepulse levels. DLM decreased ASR and TSR on trials without prepulses but not on trials with acoustic prepulses. DLM also decreased ASR when light prepulses were present 4 h post-treatment. A final experiment assessed whether the house light in the test cabinet affected ASR and TSR after PRM or DLM exposure. Rats had increased ASR and TSR when house lights were on compared with when they were off, but lighting did not differentially interact with PRM or DLM. Light and acoustic prepulses of ASR and TSR have different effects depending on the test agent and the test parameters.


Asunto(s)
Estimulación Acústica/efectos adversos , Nitrilos/farmacología , Permetrina/farmacología , Estimulación Física/efectos adversos , Inhibición Prepulso/efectos de los fármacos , Piretrinas/farmacología , Reflejo de Sobresalto/efectos de los fármacos , Estimulación Acústica/métodos , Factores de Edad , Animales , Femenino , Insecticidas/farmacología , Masculino , Estimulación Física/métodos , Inhibición Prepulso/fisiología , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/fisiología
6.
Eur J Pharmacol ; 897: 173949, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33607108

RESUMEN

Schizophrenia is a devastating neurodevelopmental disorder. The animal model based on perinatal immune activation, as first-hit, combined with peripubertal stress, as a second hit, has gained evidence in recent years. Omega-3 polyunsaturated fatty acids (n3-PUFAs) is being a promise for schizophrenia prevention. Nevertheless, the influence of sex in schizophrenia neurobiology and prevention has been neglected. Thus, the present study evaluates the preventive effects of n3-PUFAs in both sexes' mice submitted to the two-hit model and the participation of oxidative changes in this mechanism. The two-hit consisted of polyI:C administration from postnatal days (PNs) 5-7, and unpredictable stress from PNs35-43. n3-PUFAs were administered from PNs30-60. Prepulse inhibition of the startle reflex (PPI), social interaction, and Y-maze tests were conducted between PNs70-72 to evaluate positive-, negative-, and cognitive-like schizophrenia symptoms. We assessed brain oxidative changes in brain areas and plasma. Both sexes' two-hit mice presented deficits in PPI, social interaction, and working memory that were prevented by n3-PUFAs. In two-hit females, n3-PUFAs prevented increments in nitrite levels in the prefrontal cortex (PFC), hippocampus, striatum, and plasma TBARS levels. In two-hit males, n3-PUFAs prevented the increase in TBARS in the PFC, hippocampus, and striatum. Notably, male mice that received only n3-PUFAs without hit exposure presented impairments in working memory and social interaction. These results add further preclinical evidence for n3-PUFAs as an accessible and effective alternative in preventing behavioral and oxidative changes related to schizophrenia but call attention to the need for precaution in this indication due to hit- and sex-sensitive issues.


Asunto(s)
Antioxidantes/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Estrés Oxidativo/efectos de los fármacos , Esquizofrenia/prevención & control , Psicología del Esquizofrénico , Factores de Edad , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Poli I-C , Inhibición Prepulso/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Esquizofrenia/etiología , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Factores Sexuales , Desarrollo Sexual , Conducta Social , Estrés Psicológico/complicaciones
7.
Neuroreport ; 31(16): 1128-1133, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-32956214

RESUMEN

Increasing evidence supports schizophrenia may be a neurodevelopmental and neurodegenerative disorder. Fluoxetine, a selective serotonin reuptake inhibitor, has been reported to have neuroprotective effects and be effective in treating neurodegenerative disorders including schizophrenia. The objective of the present study was to evaluate the effect and underlying neuroprotective mechanism of fluoxetine on the sensorimotor gating deficit, a schizophrenia-like behavior in a neurodevelopmental schizophrenic mouse model induced by MK-801, an N-methyl-D-aspartate glutamate receptor antagonist. On postnatal day 7, mouse pups were treated with a total seven subcutaneous daily injections of MK-801 (1 mg/kg/day), followed by intraperitoneal injection of fluoxetine (5 or 10 mg/kg/day) starting on postnatal day 14 in the MK-801-injected mice for 4 weeks. The sensorimotor gating deficit in mice was measured by prepulse inhibition (PPI) behavioral test on postnatal day 43. After the behavioral test, the protein expression of brain-derived neurotrophic factor (BDNF) was measured by western blot or ELISA in the frontal cortex of mice. Our results showed fluoxetine attenuated PPI deficit and the decrease of cerebral BDNF expression in the MK-801-injected mice. These results suggest that fluoxetine can be used to treat sensorimotor gating deficit in a neurodevelopmental mouse model of schizophrenia, and the attenuating effect of fluoxetine on sensorimotor gating deficit may be related to fluoxetine's neuroprotective effect targeting on the modulation of cerebral BDNF.


Asunto(s)
Maleato de Dizocilpina/toxicidad , Antagonistas de Aminoácidos Excitadores/toxicidad , Fluoxetina/farmacología , Inhibición Prepulso/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estimulación Acústica/efectos adversos , Estimulación Acústica/métodos , Animales , Animales Recién Nacidos , Femenino , Ratones , Ratones Endogámicos ICR , Inhibición Prepulso/fisiología , Reflejo de Sobresalto/fisiología
8.
Neuropharmacology ; 179: 108275, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32835765

RESUMEN

The amygdala is a key component of the neural circuits mediating the processing and response to emotionally salient stimuli. Amygdala lesions dysregulate social interactions, responses to fearful stimuli, and autonomic functions. In rodents, the basolateral and central nuclei of the amygdala have divergent roles in behavioral control. However, few studies have selectively examined these nuclei in the primate brain. Moreover, the majority of non-human primate studies have employed lesions, which only allow for unidirectional manipulation of amygdala activity. Thus, the effects of amygdala disinhibition on behavior in the primate are unknown. To address this gap, we pharmacologically inhibited by muscimol or disinhibited by bicuculline methiodide the basolateral complex of the amygdala (BLA; lateral, basal, and accessory basal) in nine awake, behaving male rhesus macaques (Macaca mulatta). We examined the effects of amygdala manipulation on: (1) behavioral responses to taxidermy snakes and social stimuli, (2) food competition and social interaction in dyads, (3) autonomic arousal as measured by cardiovascular response, and (4) prepulse inhibition of the acoustic startle (PPI) response. All modalities were impacted by pharmacological inhibition and/or disinhibition. Amygdala inhibition decreased fear responses to snake stimuli, increased examination of social stimuli, reduced competitive reward-seeking in dominant animals, decreased heart rate, and increased PPI response. Amygdala disinhibition restored fearful response after habituation to snakes, reduced competitive reward-seeking behavior in dominant animals, and lowered heart rate. Thus, both hypoactivity and hyperactivity of the basolateral amygdala can lead to dysregulated behavior, suggesting that a narrow range of activity is necessary for normal functions.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Emociones/efectos de los fármacos , Agonistas de Receptores de GABA-A/administración & dosificación , Antagonistas de Receptores de GABA-A/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Interacción Social/efectos de los fármacos , Estimulación Acústica/métodos , Amígdala del Cerebelo/diagnóstico por imagen , Amígdala del Cerebelo/fisiología , Animales , Emociones/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Miedo/psicología , Frecuencia Cardíaca/fisiología , Inyecciones Intraventriculares , Macaca mulatta , Masculino , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Serpientes
9.
Psychopharmacology (Berl) ; 237(10): 2993-3006, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32594186

RESUMEN

RATIONALE: There are controversial reports on the effects of gabapentin in respect to psychotic symptoms. Prepulse inhibition of the acoustic startle response is an operational measure of sensorimotor gating. In laboratory rodents, deficits in sensorimotor gating are used to model behavioral endophenotypes of schizophrenia. Sleep deprivation disrupts prepulse inhibition and can be used as a psychosis model to evaluate effects of gabapentin. OBJECTIVES: This study aimed to investigate behavioral effects of gabapentin in both naïve and sleep-deprived rats. METHODS: Sleep deprivation was induced in male Wistar rats by using the modified multiple platform technique in a water tank for 72 h. The effect of water tank itself was studied in a sham group. The effects of oral acute and subchronic (4.5 days) gabapentin doses (25, 100, or 200 mg/kg/day) on sensorimotor gating and locomotor activity was evaluated by prepulse inhibition test and locomotor activity test, respectively. Plasma gabapentin levels of some groups and body weights of all groups were also assessed. RESULTS: Sleep deprivation disrupted prepulse inhibition, increased locomotor activity, reduced gabapentin plasma levels, and body weights. Some gabapentin doses disrupted sensorimotor gating irrespective of sleep condition. Some gabapentin doses increased locomotor activity in non-sleep-deprived rats and decreased locomotor activity in sleep-deprived rats. On the contrary, gabapentin did not normalize sleep deprivation-induced disruption in sensorimotor gating. CONCLUSIONS: Sleep deprivation via modified multiple platform technique could be used as an animal model for psychosis. Gabapentin may have dose- and duration-dependent effects on sensorimotor gating and locomotor activity.


Asunto(s)
Estimulación Acústica/efectos adversos , Ansiolíticos/uso terapéutico , Gabapentina/uso terapéutico , Inhibición Prepulso/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Privación de Sueño/tratamiento farmacológico , Animales , Ansiolíticos/farmacología , Relación Dosis-Respuesta a Droga , Gabapentina/farmacología , Masculino , Inhibición Prepulso/fisiología , Ratas , Ratas Wistar , Reflejo de Sobresalto/fisiología , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/fisiología , Privación de Sueño/fisiopatología , Privación de Sueño/psicología
10.
Behav Brain Res ; 376: 112077, 2019 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-31499090

RESUMEN

Rivastigmine (RVT) is a reversible inhibitor of cholinesterase approved worldwide for the treatment of cognitive dysfunctions, especially in Alzheimer's disease. Most previous pre-clinical studies have examined the effects of RVT treatment in a wide variety of pathological research models. Nonetheless, the effects of this drug on sensorimotor gating, memory, and learning tasks in healthy subjects remains unclear. In this study, we investigate the procognitive effects of RVT treatment in healthy rats through sensorimotor gating evaluations (measured as prepulse inhibition of the acoustic startle reflex), active avoidance learning, and spatial memory learning in a radial maze. There is an increase in the amplitude of the startle reflex in RVT-treated rats compared to the control groups, whereas the latency remained constant. Sensorimotor gating values were also incremented compared to those values from controls. In active avoidance, rats treated with RVT learned faster to successfully perform the task compared to controls, but afterwards all groups exhibited virtually identical results. During the sessions in the radial maze, RVT-treated rats committed fewer errors in both the working and reference memory compared to controls. All in all, our results support the hypothesis that RVT treatment may entail procognitive effects in healthy subjects.


Asunto(s)
Reflejo de Sobresalto/fisiología , Rivastigmina/farmacología , Corteza Sensoriomotora/efectos de los fármacos , Estimulación Acústica , Animales , Reacción de Prevención/fisiología , Encéfalo/fisiología , Inhibidores de la Colinesterasa/metabolismo , Inhibidores de la Colinesterasa/farmacología , Colinesterasas/metabolismo , Cognición/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Wistar , Reflejo de Sobresalto/efectos de los fármacos , Rivastigmina/metabolismo , Filtrado Sensorial/fisiología , Aprendizaje Espacial/fisiología , Memoria Espacial/fisiología
11.
J Ethnopharmacol ; 235: 320-328, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-30776471

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: The plant arctic root (Rhodiola rosea, L.) is growing in northern regions of Europe, Asia and North America. Extracts of R. rosea are used in traditional medicine for various conditions related to nervous system function. According to scientific studies from the last decades, the plant might have potential for use in the treatment of memory impairments, stress and depression, but reports concerning other neuropsychiatric disorders are scarce. AIM OF THE STUDY: In this context, our study aimed to examine potential antipsychotic-like effects of R. rosea root extract. MATERIALS AND METHODS: We tested the effects of R. rosea root extract on prepulse inhibition in rats and mice. Prepulse inhibition is an established operational measure of sensorimotor gating, which is impaired in schizophrenia and other psychotic disorders. RESULTS: R. rosea root extract increased prepulse inhibition in rats and mice. Interestingly, the R. rosea extract had stronger effects in those individual animals that had low baseline levels of prepulse inhibition. Therefore, we performed further experiments in which we pharmacologically induced a prepulse inhibition deficit by two different psychostimulants, either the dopamine D2 receptor agonist apomorphine or the NMDA receptor antagonist dizocilpine (MK-801). Pre-treatment with the R. rosea extract significantly restored both, apomorphine- and dizocilpine-induced prepulse inhibition deficits. CONCLUSIONS: The present study demonstrates that R. rosea extract robustly reverses prepulse inhibition deficits in rodents. This suggests antipsychotic-like effects of R. rosea extract. Future studies should focus on the pharmacological mechanisms underlying these effects.


Asunto(s)
Antipsicóticos/farmacología , Extractos Vegetales/farmacología , Rhodiola/química , Filtrado Sensorial/efectos de los fármacos , Animales , Antipsicóticos/aislamiento & purificación , Apomorfina/administración & dosificación , Modelos Animales de Enfermedad , Maleato de Dizocilpina/administración & dosificación , Masculino , Medicina Tradicional/métodos , Ratones , Ratones Endogámicos C57BL , Raíces de Plantas , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Wistar
12.
Behav Brain Res ; 360: 312-322, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30521932

RESUMEN

Early life immune challenges are risk factors for neurodevelopmental disorders. In adolescence, they elicit behavioral symptoms that resemble clinical disorders. Stressors during this time may alter signaling from the gut microbiome, which increases the risk for psychiatric disorders. It was hypothesized that adolescent immune challenges may interact with a gut bacterial product, the short-chain fatty acid, propionic acid (PPA), to potentiate symptoms of anxiety and sensory abnormality. The present study investigated the effects of repeated lipopolysaccharide (LPS) exposure during early adolescence, on the behavioral effects of PPA in late adolescence and adulthood. Male adolescent rats were injected with LPS (0.2 mg/kg i.p.) or the vehicle on postnatal days (P) 28, P30, P32, and P34. They were later administered either PPA (500 mg/kg i.p.) or the vehicle during late adolescence on P40 and P43, and were subsequently tested on the light-dark anxiety test and acoustic startle response, respectively. In adulthood, the rats were again injected with PPA or the vehicle and tested on the light-dark and acoustic startle tasks on P74 and P77. The results of this study showed that LPS and PPA both decreased locomotor activity. PPA reduced vertical activity, percent prepulse inhibition, and acoustic startle response magnitude. LPS increased anxiogenic behaviors and induced a delayed increase in acoustic startle response magnitude in adulthood. Although no LPS and PPA interactions were found, the results of this study suggest that early adolescent immune activation can induce long-term behavioral changes that resemble the complex phenotypes of clinical disorders.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Conducta de Enfermedad/efectos de los fármacos , Lipopolisacáridos/farmacología , Propionatos/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Estimulación Acústica , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Adaptación a la Oscuridad/efectos de los fármacos , Locomoción/efectos de los fármacos , Estudios Longitudinales , Masculino , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Long-Evans
13.
Mol Neurobiol ; 56(6): 4492-4517, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30338483

RESUMEN

We utilized a cell-level approach to examine glycolytic pathways in the DLPFC of subjects with schizophrenia (n = 16) and control (n = 16) and found decreased mRNA expression of glycolytic enzymes in pyramidal neurons, but not astrocytes. To replicate these novel bioenergetic findings, we probed independent datasets for bioenergetic targets and found similar abnormalities. Next, we used a novel strategy to build a schizophrenia bioenergetic profile by a tailored application of the Library of Integrated Network-Based Cellular Signatures data portal (iLINCS) and investigated connected cellular pathways, kinases, and transcription factors using Enrichr. Finally, with the goal of identifying drugs capable of "reversing" the bioenergetic schizophrenia signature, we performed a connectivity analysis with iLINCS and identified peroxisome proliferator-activated receptor (PPAR) agonists as promising therapeutic targets. We administered a PPAR agonist to the GluN1 knockdown model of schizophrenia and found it improved long-term memory. Taken together, our findings suggest that tailored bioinformatics approaches, coupled with the LINCS library of transcriptional signatures of chemical and genetic perturbagens, may be employed to identify novel treatment strategies for schizophrenia and related diseases.


Asunto(s)
Metabolismo Energético , Redes Reguladoras de Genes , Esquizofrenia/metabolismo , Esquizofrenia/terapia , Animales , Análisis por Conglomerados , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Descubrimiento de Drogas , Metabolismo Energético/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Captura por Microdisección con Láser , Masculino , Ratones , Actividad Motora/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Pioglitazona/farmacología , Inhibición Prepulso/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Reproducibilidad de los Resultados , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Conducta Estereotipada/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Schizophr Res ; 198: 6-15, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28549722

RESUMEN

BACKGROUND: The Consortium on the Genetics of Schizophrenia (COGS) collected case-control endophenotype and genetic information from 2457 patients and healthy subjects (HS) across 5 test sites over 3.5 years. Analysis of the first "wave" (W1) of 1400 subjects identified prepulse inhibition (PPI) deficits in patients vs. HS. Data from the second COGS "wave" (W2), and the combined W(1+2), were used to assess: 1) the replicability of PPI deficits in this design; 2) the impact of response criteria on PPI deficits; and 3) PPI in a large cohort of antipsychotic-free patients. METHODS: PPI in W2 HS (n=315) and schizophrenia patients (n=326) was compared to findings from W1; planned analyses assessed the impact of diagnosis, "wave" (1 vs. 2), and startle magnitude criteria. Combining waves allowed us to assess PPI in 120 antipsychotic-free patients, including many in the early course of illness. RESULTS: ANOVA of all W(1+2) subjects revealed robust PPI deficits in patients across "waves" (p<0.0004). Strict response criteria excluded almost 39% of all subjects, disproportionately impacting specific subgroups; ANOVA in this smaller cohort confirmed no significant effect of "wave" or "wave x diagnosis" interaction, and a significant effect of diagnosis (p<0.002). Antipsychotic-free, early-illness patients had particularly robust PPI deficits. DISCUSSION: Schizophrenia-linked PPI deficits were replicable across two multi-site "waves" of subjects collected over 3.5years. Strict response criteria disproportionately excluded older, male, non-Caucasian patients with low-normal hearing acuity. These findings set the stage for genetic analyses of PPI using the combined COGS wave 1 and 2 cohorts.


Asunto(s)
Trastornos Neurológicos de la Marcha/etiología , Inhibición Neural/fisiología , Inhibición Prepulso/fisiología , Esquizofrenia/complicaciones , Estimulación Acústica , Adolescente , Adulto , Análisis de Varianza , Antipsicóticos/uso terapéutico , Estudios de Cohortes , Endofenotipos , Femenino , Trastornos Neurológicos de la Marcha/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Inhibición Neural/efectos de los fármacos , Inhibición Prepulso/efectos de los fármacos , Escalas de Valoración Psiquiátrica , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/epidemiología , Adulto Joven
15.
Schizophr Res ; 198: 60-67, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28864281

RESUMEN

The Brain-Derived Neurotrophic Factor (BDNF) Val66Met polymorphism results in reduced activity-dependent BDNF release and has been implicated in schizophrenia. However, effects of the polymorphism on functional dopaminergic and N-methyl-d-aspartate (NMDA) receptor-associated activity remain unclear. We used prepulse inhibition, a measure of sensorimotor gating which is disrupted in schizophrenia, and assessed the effects of acute treatment with the dopamine receptor agonist, apomorphine (APO), and the NMDA receptor antagonist, MK-801. We used adult humanized hBDNFVal66Met 'knockin' mice which express either the Val/Val, Val/Met or Met/Met genotype. An interaction of BDNF with stress was modelled by chronic young-adult treatment with corticosterone (CORT). At 1 or 3mg/kg, APO had no effect in Val/Val mice but significantly reduced PPI at the 100ms inter-stimulus interval (ISI) in Val/Met and Met/Met mice. However, after CORT pretreatment, APO significantly reduced PPI in all genotypes similarly. At 0.1 or 0.25mg/kg, MK-801 significantly disrupted PPI at the 100ms ISI independent of genotype or CORT pretreatment. There were differential effects of APO and MK-801 on PPI at the 30ms ISI and startle between the genotypes, irrespective of CORT pretreatment. These results show that the BDNF Val66Met Val/Met and Met/Met genotypes are more sensitive than the Val/Val genotype to the effect of APO on PPI. A history of stress, here modelled by chronic CORT administration, increases effects of APO in Val/Val mice.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Polimorfismo Genético/genética , Inhibición Prepulso/genética , Estrés Psicológico/genética , Estrés Psicológico/fisiopatología , Estimulación Acústica , Análisis de Varianza , Animales , Apomorfina/farmacología , Apomorfina/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corticosterona/administración & dosificación , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Maleato de Dizocilpina/uso terapéutico , Agonistas de Dopamina/farmacología , Agonistas de Dopamina/uso terapéutico , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Genotipo , Humanos , Metionina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Prepulso/efectos de los fármacos , Valina/genética
16.
Psychopharmacology (Berl) ; 234(18): 2727-2735, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28674745

RESUMEN

RATIONALE: Altered glutamate NMDA receptor function is implicated in schizophrenia, and gender differences have been demonstrated in this illness. OBJECTIVES: This study aimed to investigate the interaction of gonadal hormones with NMDA receptor-mediated locomotor hyperactivity and PPI disruption in mice. RESULTS: The effect of 0.25 mg/kg of MK-801 on locomotor activity was greater in male mice than in female mice. Gonadectomy (by surgical castration) significantly reduced MK-801-induced hyperlocomotion in male mice, but no effect of gonadectomy was seen in female mice or on amphetamine-induced locomotor hyperactivity. The effect of MK-801 on prepulse inhibition of startle (PPI) was similar in intact and castrated male mice and in ovariectomized (OVX) female mice. In contrast, there was no effect of MK-801 on PPI in intact female mice. Forebrain NMDA receptor density, as measured with [3H]MK-801 autoradiography, was significantly higher in male than in female mice but was not significantly altered by either castration or OVX. CONCLUSIONS: These results suggest that male sex hormones enhance the effect of NMDA receptor blockade on psychosis-like behaviour. This interaction was not seen in female mice and was independent of NMDA receptor density in the forebrain. Male sex hormones may be involved in psychosis by an interaction with NMDA receptor hypofunction.


Asunto(s)
Hormonas Esteroides Gonadales/metabolismo , Hipercinesia/inducido químicamente , Hipercinesia/metabolismo , Receptores de N-Metil-D-Aspartato/fisiología , Estimulación Acústica/métodos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Maleato de Dizocilpina/toxicidad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Orquiectomía , Ovariectomía , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología
17.
eNeuro ; 4(1)2017.
Artículo en Inglés | MEDLINE | ID: mdl-28275719

RESUMEN

New antidepressant pharmacotherapies that provide rapid relief of depressive symptoms are needed. The NMDA receptor antagonist ketamine exerts rapid antidepressant actions in depressed patients but also side effects that complicate its clinical utility. Ketamine promotes excitatory synaptic strength, likely by producing high-frequency correlated activity in mood-relevant regions of the forebrain. Negative allosteric modulators of GABA-A receptors containing α5 subunits (α5 GABA-NAMs) should also promote high-frequency correlated electroencephalogram (EEG) activity and should therefore exert rapid antidepressant responses. Because α5 subunits display a restricted expression in the forebrain, we predicted that α5 GABA-NAMs would produce activation of principle neurons but exert fewer side effects than ketamine. We tested this hypothesis in male mice and observed that the α5 GABA-NAM MRK-016 exerted an antidepressant-like response in the forced swim test at 1 and 24 h after administration and an anti-anhedonic response after chronic stress in the female urine sniffing test (FUST). Like ketamine, MRK-016 produced a transient increase in EEG γ power, and both the increase in γ power and its antidepressant effects in the forced swim test were blocked by prior administration of the AMPA-type glutamate receptor antagonist 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX). Unlike ketamine, however, MRK-016 produced no impairment of rota-rod performance, no reduction of prepulse inhibition (PPI), no conditioned-place preference (CPP), and no change in locomotion. α5 GABA-NAMs, thus reproduce the rapid antidepressant-like actions of ketamine, perhaps via an AMPA receptor (AMPAR)-dependent increase in coherent neuronal activity, but display fewer potential negative side effects. These compounds thus demonstrate promise as clinically useful fast-acting antidepressants.


Asunto(s)
Antidepresivos/farmacología , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Isoxazoles/farmacología , Receptores de GABA-A/metabolismo , Triazinas/farmacología , Regulación Alostérica , Animales , Condicionamiento Psicológico/efectos de los fármacos , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/fisiopatología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ritmo Gamma/efectos de los fármacos , Ketamina/efectos adversos , Ketamina/farmacología , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Inhibición Prepulso/efectos de los fármacos , Quinoxalinas/farmacología , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo
18.
Neuropsychopharmacology ; 42(11): 2163-2177, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28294132

RESUMEN

All FDA-approved antipsychotic drugs (APDs) target primarily dopamine D2 or serotonin (5-HT2A) receptors, or both; however, these medications are not universally effective, they may produce undesirable side effects, and provide only partial amelioration of negative and cognitive symptoms. The heterogeneity of pharmacological responses in schizophrenic patients suggests that additional drug targets may be effective in improving aspects of this syndrome. Recent evidence suggests that 5-HT2C receptors may be a promising target for schizophrenia since their activation reduces mesolimbic nigrostriatal dopamine release (which conveys antipsychotic action), they are expressed almost exclusively in CNS, and have weight-loss-promoting capabilities. A difficulty in developing 5-HT2C agonists is that most ligands also possess 5-HT2B and/or 5-HT2A activities. We have developed selective 5-HT2C ligands and herein describe their preclinical effectiveness for treating schizophrenia-like behaviors. JJ-3-45, JJ-3-42, and JJ-5-34 reduced amphetamine-stimulated hyperlocomotion, restored amphetamine-disrupted prepulse inhibition, improved social behavior, and novel object recognition memory in NMDA receptor hypofunctioning NR1-knockdown mice, and were essentially devoid of catalepsy. However, they decreased motivation in a breakpoint assay and did not promote reversal learning in MK-801-treated mice. Somewhat similar effects were observed with lorcaserin, a 5-HT2C agonist with potent 5-HT2B and 5-HT2A agonist activities, which is approved for treating obesity. Microdialysis studies revealed that both JJ-3-42 and lorcaserin reduced dopamine efflux in the infralimbic cortex, while only JJ-3-42 decreased it in striatum. Collectively, these results provide additional evidence that 5-HT2C receptors are suitable drug targets with fewer side effects, greater therapeutic selectivity, and enhanced efficacy for treating schizophrenia and related disorders than current APDs.


Asunto(s)
Inhibición Prepulso/efectos de los fármacos , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Agonistas del Receptor de Serotonina 5-HT2/uso terapéutico , Estimulación Acústica/efectos adversos , Anfetamina/toxicidad , Animales , Benzazepinas/farmacología , Catalepsia/tratamiento farmacológico , Catalepsia/etiología , Aprendizaje Discriminativo/efectos de los fármacos , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Conducta Exploratoria/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Motivación/efectos de los fármacos , Neurotransmisores/metabolismo , Reconocimiento en Psicología/efectos de los fármacos , Esquizofrenia/inducido químicamente , Agonistas del Receptor de Serotonina 5-HT2/química , Conducta Social
19.
J Psychopharmacol ; 31(4): 505-513, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28114835

RESUMEN

Prepulse inhibition (PPI) is a behavioral test in which the startle reflex response to a high-intensity stimulus (pulse) is inhibited by the prior presentation of a weak stimulus (prepulse). The classic neural circuitry that mediates startle response is localized in the brainstem; however, recent studies point to the contribution of structures involved in higher cognitive functions in regulating the sensorimotor gating, particularly forebrain regions innervated by dopaminergic nuclei. The aim of the present study was to verify the role of dorsal striatum (DS) and dopaminergic transmitting mediated by D1 and D2 receptors on PPI test in rats. DS inactivation induced by muscimol injection did not affect PPI (%PPI and startle response), although it impaired the locomotor activity and caused catalepsy. Infusion of D1-like antagonist SCH23390 impaired %PPI but did not disturb the startle response and locomotor activity evaluated immediately after PPI test. D2 antagonist microinjection (sulpiride) did not affect %PPI and startle response, but impaired motor activity. These results point to an important role of DS, probably mediated by direct basal ganglia pathway, on modulation of sensorimotor gating, in accordance with clinical studies showing PPI deficits in schizophrenia, Tourette syndrome, and compulsive disorders - pathologies related to basal ganglia dysfunctions.


Asunto(s)
Neuronas/metabolismo , Inhibición Prepulso/fisiología , Receptores de Dopamina D1/metabolismo , Filtrado Sensorial/fisiología , Asta Dorsal de la Médula Espinal/metabolismo , Estimulación Acústica/métodos , Animales , Benzazepinas/farmacología , Dopamina/metabolismo , Antagonistas de Dopamina/farmacología , Masculino , Inhibición Neural/efectos de los fármacos , Inhibición Neural/fisiología , Neuronas/efectos de los fármacos , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Dopamina D2/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Filtrado Sensorial/efectos de los fármacos , Asta Dorsal de la Médula Espinal/efectos de los fármacos
20.
Behav Brain Res ; 320: 517-525, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27725171

RESUMEN

Although N-methyl-d-aspartate receptor antagonists-induced hypoglutamate rodent models are the most well-established models for preclinical studies of schizophrenia-related deficits, they also evoke a wide spectrum of psychotomimetic side effects. It is significant to increase the specificity of hypoglutamate rodent models. In this study, the recognition memory was evaluated in rats by object recognition test (ORT), sensorimotor gating was evaluated by prepulse inhibition of the startle reflex (PPI), and locomotor activity was measured using open field test. High-performance liquid chromatography was used to measure neurotransmitters content in the medial prefrontal cortex (mPFC) and thalamus (THA). Total Akt and phospho-Akt protein was measured by Western blots. Results showed that 0.3mg/kg of MK-801 was most effective in inducing locomotion. 0.3mg/kg of MK-801 was most effective in decreasing PPI. 0.03mg/kg of MK-801 was most effective in decreasing object memory while not affecting exploration manners in the training session. 0.03mg/kg of MK-801 significantly increased HVA and Glu content in the mPFC. 0.1mg/kg of MK-801 significantly decreased GABA content in the THA. 0.03mg/kg of MK-801 significantly decreased Akt phosphorylation in the mPFC, which was related to the ORT index. In conclusion, a dose of 0.03mg/kg MK-801 can establish a "pure" memory impairment model without contaminations of sensorimotor gating and locomotor activity. MK-801-induced cognitive deficits is associated with increased DA metabolites and glutamate content in the mPFC and decreased GABA content in the THA as well as decrease in Akt phosphorylation in the mPFC.


Asunto(s)
Modelos Animales de Enfermedad , Maleato de Dizocilpina/toxicidad , Antagonistas de Aminoácidos Excitadores/toxicidad , Trastornos de la Memoria/inducido químicamente , Aminoácidos/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Locomoción/efectos de los fármacos , Masculino , Neurotransmisores/metabolismo , Proteína Oncogénica v-akt/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Inhibición Prepulso/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA