Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuropsychopharmacology ; 48(8): 1155-1163, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36400921

RESUMEN

ADHD is a highly prevalent neurodevelopmental disorder. The first-line therapeutic for ADHD, methylphenidate, can cause serious side effects including weight loss, insomnia, and hypertension. Therefore, the development of non-stimulant-based therapeutics has been prioritized. However, many of these also cause other effects, most notably somnolence. Here, we have used a uniquely powerful genetic model and unbiased drug screen to identify novel ADHD non-stimulant therapeutics. We first found that adgrl3.1 null (adgrl3.1-/-) zebrafish larvae showed a robust hyperactive phenotype. Although the hyperactivity was rescued by three ADHD non-stimulant therapeutics, all interfered significantly with sleep. Second, we used wild-type zebrafish larvae to characterize a simple behavioral phenotype generated by atomoxetine and screened the 1200 compound Prestwick Chemical Library® for a matching behavioral profile resulting in 67 hits. These hits were re-assayed in the adgrl3.1-/-. Using the previously identified non-stimulants as a positive control, we identified four compounds that matched the effect of atomoxetine: aceclofenac, amlodipine, doxazosin, and moxonidine. We additionally demonstrated cognitive effects of moxonidine in mice using a T-maze spontaneous alternation task. Moxonidine, has high affinity for imidazoline 1 receptors. We, therefore, assayed a pure imidazoline 1 agonist, LNP599, which generated an effect closely matching other non-stimulant ADHD therapeutics suggesting a role for this receptor system in ADHD. In summary, we introduce a genetic model of ADHD in zebrafish and identify five putative therapeutics. The findings offer a novel tool for understanding the neural circuits of ADHD, suggest a novel mechanism for its etiology, and identify novel therapeutics.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Estimulantes del Sistema Nervioso Central , Imidazolinas , Metilfenidato , Animales , Ratones , Clorhidrato de Atomoxetina/farmacología , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Trastorno por Déficit de Atención con Hiperactividad/genética , Pez Cebra , Metilfenidato/farmacología , Metilfenidato/uso terapéutico , Fenotipo , Imidazolinas/uso terapéutico , Estimulantes del Sistema Nervioso Central/efectos adversos
2.
PLoS One ; 17(1): e0259753, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34986152

RESUMEN

In the present study, we characterize a novel zebrafish mutant of solute carrier 18A2 (slc18a2), also known as vesicular monoamine transporter 2 (vmat2), that exhibits a behavioural phenotype partially consistent with human Parkinson´s disease. At six days-post-fertilization, behaviour was analysed and demonstrated that vmat2 homozygous mutant larvae, relative to wild types, show changes in motility in a photomotor assay, altered sleep parameters, and reduced dopamine cell number. Following an abrupt lights-off stimulus mutant larvae initiate larger movements but subsequently inhibit them to a lesser extent in comparison to wild-type larvae. Conversely, during a lights-on period, the mutant larvae are hypomotile. Thigmotaxis, a preference to avoid the centre of a behavioural arena, was increased in homozygotes over heterozygotes and wild types, as was daytime sleep ratio. Furthermore, incubating mutant larvae in pramipexole or L-Dopa partially rescued the motor phenotypes, as did injecting glial cell-derived neurotrophic factor (GDNF) into their brains. This novel vmat2 model represents a tool for high throughput pharmaceutical screens for novel therapeutics, in particular those that increase monoamine transport, and for studies of the function of monoamine transporters.


Asunto(s)
Locomoción/genética , Proteínas de Transporte Vesicular de Monoaminas/genética , Proteínas de Transporte Vesicular de Monoaminas/metabolismo , Animales , Encéfalo/metabolismo , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
3.
Front Behav Neurosci ; 14: 623924, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33390914

RESUMEN

Parkinson's disease (PD) has been modeled in several animal species using the neurotoxins 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and its oxidized product 1-methyl-4-phenylpyridinium (MPP+). MPP+ selectively kills dopaminergic neurons in pars compacta of the substantia nigra, inducing parkinsonian symptoms in animals. Typically, neurotoxicity models of PD in zebrafish assess acute drug effects on locomotion. In the present study, we examined the lasting effects of MPP+ exposure and drug treatment in zebrafish larvae. Larvae were incubated in 500 µM MPP+, from 1 to 5 days post fertilization (dpf), followed by 24 h drug-free acclimation. At 6 dpf, the behavior was analyzed for locomotion, thigmotaxis, and sleep. Next, in separate assays we assessed the drug effects of brain injected glial cell-derived neurotrophic factor (GDNF) and 4-phenylbutyrate (PBA), co-incubated with MPP+. We show that MPP+ exposure consistently reduces swim distance, movement frequency, and cumulative time of movement; thus mimicking a parkinsonian phenotype of reduced movement. In contrast, MPP+ exposed larvae demonstrate reduced anxiety-like behavior and exhibit a sleep phenotype inconsistent with human PD: the larvae display longer sleep bouts, less sleep fragmentation, and more sleep. Previously reported rescuing effects of PBA were not replicated in this study. Moreover, whereas GDNF attenuated the sleep phenotype induced by MPP+, PBA augmented it. The current data suggest that MPP+ exposure generates a multifaceted phenotype in zebrafish and highlights that analyzing a narrow window of data can reveal effects that may be inconsistent with longer multi-parameter approaches. It further indicates that the model generally captures motor symptoms more faithfully than non-motor symptoms.

4.
Psychiatry Res ; 212(3): 223-9, 2013 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-23149021

RESUMEN

Emotional stimuli may draw attention to such an extent that they hamper the processing of subsequent signals, a phenomenon termed emotion-induced blindness (EIB). As depersonalization is associated with self-reported attenuated emotional responses, the present study explored whether individuals scoring high on the Cambridge Depersonalization Scale (CDS; n=15) exhibit a diminished EIB effect relative to low CDS scoring individuals (n=15), and whether attentional processes reflected in event-related potentials (ERPs) are implicated in this effect. We obtained an EIB effect such that emotional distractors that preceded targets with a lag of 200ms reduced correct detection of targets. Although the magnitude of this effect was similar for high and low CDS participants, high CDS participants exhibited a significantly lower ERP amplitude at the frontal lead in the 200-300ms window than did low CDS individuals to targets that followed emotional versus neutral distractors. This latter effect was significantly related to the Alienation factor of the CDS. This pattern suggests that difficulties in the discrimination between emotional and neutral stimuli relate to the feeling of unreality in depersonalization.


Asunto(s)
Encéfalo/fisiopatología , Despersonalización/fisiopatología , Emociones/fisiología , Potenciales Evocados/fisiología , Atención/fisiología , Mapeo Encefálico , Señales (Psicología) , Despersonalización/psicología , Discriminación en Psicología/fisiología , Electroencefalografía , Femenino , Humanos , Masculino , Autoinforme , Adulto Joven
5.
Zebrafish ; 8(3): 133-40, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21882999

RESUMEN

We describe, for the first time, the effects of the wakefulness-promoting drug modafinil on sleep and wakefulness in larval zebrafish. Modafinil is currently used to treat narcolepsy, hypersomnia, and shift-work disorder by increasing wakefulness. Tolerance and dependence are limited with modafinil use, differentiating it from common stimulants; however, the neural mechanisms of action of modafinil are still unknown. Zebrafish, a low-cost, prolific, and genetically tractable animal model, have recently become a key model in sleep research. Zebrafish express circadian rhythms, sleep homeostasis, and sleep pressure, and, in addition, respond to common hypnotics and stimulants in a manner similar to mammals. Therefore, in the current experiment we characterize the effects of modafinil on sleep-wake cycles in larval zebrafish as a first step to gaining further insight into the neural mechanisms underlying the effects of modafinil. We show that modafinil modulates sleep-wake activity in larval zebrafish in a manner consistent with what would be predicted from mammalian data. Modafinil increases wakefulness by lengthening wake-bouts, an effect that likely restricted to the night (lights-off). These results validate the use of zebrafish as an animal model for the study of sleep and provide a means for dissecting the neural mechanisms of modafinil, and, more broadly, sleep disorders.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Ritmo Circadiano/efectos de los fármacos , Sueño/efectos de los fármacos , Vigilia/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Modafinilo , Pez Cebra
6.
Behav Brain Res ; 211(1): 11-5, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20188125

RESUMEN

The co-expression of behavioral and neural events represents a situation conducive to Hebbian-type neuroplasticity and may provide a reasonable explanation for how the amount of movement during the perinatal period contributes to neuromotor development. Direct current-coupled electrographic recordings in premature infants indicate that the majority of the electrographic activity is exhibited in a slow frequency range that is either distorted or not visible using traditional recording methods. Therefore, we provide a description of the behavioral correlates of direct current-coupled electrographic recordings in six premature human infants (3 males and 3 females; 30-34 weeks). We report, in concert with prior data, that electrographic activity and movements occur in tightly coupled discrete bouts. Surprisingly, spontaneous activity transients, which are slow, high amplitude, multiband electrographic events, typically precede startles; thereby revealing a previously unknown coupling of early neural and behavioral events in humans. Taken together, the present findings open novel venues for studying and dissecting mammalian neuromotor development.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales Evocados/fisiología , Recien Nacido Prematuro/fisiología , Actividad Motora/fisiología , Movimiento/fisiología , Nivel de Alerta/fisiología , Conducta/fisiología , Electroencefalografía , Femenino , Humanos , Recién Nacido , Masculino , Plasticidad Neuronal/fisiología , Reflejo de Sobresalto/fisiología , Factores de Tiempo
7.
Front Neurol ; 1: 156, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21212828

RESUMEN

Studies in adult mammals (rats, cats, mice, and humans) have revealed a surprising regularity in the duration of sleep and wake bouts. In particular, wake bout durations exhibit a power-law distribution whereas sleep bout durations exhibit an exponential distribution. Moreover, in rodents, sleep bouts exhibit an exponential distribution at all ages examined, whereas wake bout durations exhibit exponential distributions early in ontogeny with a clear power-law emerging only at the older ages. Thus, the data examined thus far suggests a similar developmental trajectory for a wide range of mammals which in turn may offer a novel metric to directly compare human and animal sleep-wake data. Therefore, we tested the generalizability of these findings by examining the distributions of sleep and wake bouts during the night in a healthy human sample - from premature infants to 70-year-olds. We find that sleep bouts elongate over the first years. At the same time wake bouts shorten but elongate again with increasing age. Moreover, sleep bout durations exhibit exponential distributions at all ages tested, except for the youngest (premature infants). Wake bouts exhibit a power-law distribution - but only during a restricted time window during adulthood. We conclude that the developmental trajectory of human sleep-wake cycles does not map well onto those of rodents; however, the method of characterizing sleep-wake cycles, using bout distribution, holds great promise for classifying sleep, its disorders, and tracking its developmental milestones across the lifespan in humans.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA