Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(19): e2307156121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683996

RESUMEN

Tourette disorder (TD) is poorly understood, despite affecting 1/160 children. A lack of animal models possessing construct, face, and predictive validity hinders progress in the field. We used CRISPR/Cas9 genome editing to generate mice with mutations orthologous to human de novo variants in two high-confidence Tourette genes, CELSR3 and WWC1. Mice with human mutations in Celsr3 and Wwc1 exhibit cognitive and/or sensorimotor behavioral phenotypes consistent with TD. Sensorimotor gating deficits, as measured by acoustic prepulse inhibition, occur in both male and female Celsr3 TD models. Wwc1 mice show reduced prepulse inhibition only in females. Repetitive motor behaviors, common to Celsr3 mice and more pronounced in females, include vertical rearing and grooming. Sensorimotor gating deficits and rearing are attenuated by aripiprazole, a partial agonist at dopamine type II receptors. Unsupervised machine learning reveals numerous changes to spontaneous motor behavior and less predictable patterns of movement. Continuous fixed-ratio reinforcement shows that Celsr3 TD mice have enhanced motor responding and reward learning. Electrically evoked striatal dopamine release, tested in one model, is greater. Brain development is otherwise grossly normal without signs of striatal interneuron loss. Altogether, mice expressing human mutations in high-confidence TD genes exhibit face and predictive validity. Reduced prepulse inhibition and repetitive motor behaviors are core behavioral phenotypes and are responsive to aripiprazole. Enhanced reward learning and motor responding occur alongside greater evoked dopamine release. Phenotypes can also vary by sex and show stronger affection in females, an unexpected finding considering males are more frequently affected in TD.


Asunto(s)
Dopamina , Mutación , Síndrome de Tourette , Animales , Síndrome de Tourette/genética , Síndrome de Tourette/fisiopatología , Síndrome de Tourette/metabolismo , Ratones , Femenino , Masculino , Humanos , Dopamina/metabolismo , Recompensa , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Aprendizaje/fisiología , Conducta Animal , Inhibición Prepulso/genética , Filtrado Sensorial/genética
2.
Proc Natl Acad Sci U S A ; 114(5): 1177-1182, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28096412

RESUMEN

Neurotransmission in dentate gyrus (DG) is critical for spatial coding, learning memory, and emotion processing. Although DG dysfunction is implicated in psychiatric disorders, including schizophrenia, underlying pathological mechanisms remain unclear. Here we report that transmembrane protein 108 (Tmem108), a novel schizophrenia susceptibility gene, is highly enriched in DG granule neurons and its expression increased at the postnatal period critical for DG development. Tmem108 is specifically expressed in the nervous system and enriched in the postsynaptic density fraction. Tmem108-deficient neurons form fewer and smaller spines, suggesting that Tmem108 is required for spine formation and maturation. In agreement, excitatory postsynaptic currents of DG granule neurons were decreased in Tmem108 mutant mice, indicating a hypofunction of glutamatergic activity. Further cell biological studies indicate that Tmem108 is necessary for surface expression of AMPA receptors. Tmem108-deficient mice display compromised sensorimotor gating and cognitive function. Together, these observations indicate that Tmem108 plays a critical role in regulating spine development and excitatory transmission in DG granule neurons. When Tmem108 is mutated, mice displayed excitatory/inhibitory imbalance and behavioral deficits relevant to schizophrenia, revealing potential pathophysiological mechanisms of schizophrenia.


Asunto(s)
Trastornos del Conocimiento/genética , Giro Dentado/fisiología , Filtrado Sensorial/genética , Proteínas de Transporte Vesicular/fisiología , Animales , Animales Recién Nacidos , Trastornos del Conocimiento/fisiopatología , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Electroporación , Potenciales Postsinápticos Excitadores/fisiología , Miedo , Genes Reporteros , Ácido Glutámico/fisiología , Células HEK293 , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Neuronas/fisiología , Neuronas/ultraestructura , Densidad Postsináptica/química , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores AMPA/biosíntesis , Esquizofrenia/genética , Filtrado Sensorial/fisiología , Transmisión Sináptica/fisiología , Proteínas de Transporte Vesicular/deficiencia , Proteínas de Transporte Vesicular/genética
3.
Nitric Oxide ; 80: 32-36, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30096361

RESUMEN

Research increasingly suggests that nitric oxide (NO) plays a role in the pathogenesis of schizophrenia. One important line of evidence comes from genetic studies, which have repeatedly detected an association between the neuronal isoform of nitric oxide synthase (nNOS or NOS1) and schizophrenia. However, the pathogenetic pathways linking nNOS, NO, and the disorder remain poorly understood. A deficit in sensorimotor gating is considered to importantly contribute to core schizophrenia symptoms such as psychotic disorganization and thought disturbance. We selected three candidate nNOS polymorphisms (Ex1f-VNTR, rs6490121 and rs41279104), associated with schizophrenia and cognition in previous studies, and tested their association with the efficiency of sensorimotor gating in healthy human adults. We found that risk variants of Ex1f-VNTR and rs6490121 (but not rs41279104) were associated with a weaker prepulse inhibition (PPI) of the acoustic startle reflex, a standard measure of sensorimotor gating. Furthermore, the effect of presence of risk variants in Ex1f-VNTR and rs6490121 was additive: PPI linearly decreased with increasing number of risk alleles, being highest in participants with no risk allele, while lowest in individuals who carry three risk alleles. Our findings indicate that NO is involved in the regulation of sensorimotor gating, and highlight one possible pathogenetic mechanism for NO playing a role in the development of schizophrenia psychosis.


Asunto(s)
Óxido Nítrico Sintasa de Tipo I/genética , Polimorfismo de Nucleótido Simple , Filtrado Sensorial/genética , Adulto , Exones , Femenino , Humanos , Masculino , Repeticiones de Minisatélite , Óxido Nítrico/fisiología , Inhibición Prepulso/genética , Reflejo de Sobresalto/genética , Esquizofrenia/genética
4.
Mol Psychiatry ; 21(10): 1449-59, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26728564

RESUMEN

Exploring drug targets based on disease-associated molecular mechanisms during development is crucial for the generation of novel prevention and treatment strategies for neurodevelopmental psychiatric conditions. We report that prefrontal cortex (PFC)-specific postnatal knockdown of DISC1 via in utero electroporation combined with an inducible knockdown expression system drives deficits in synaptic GABAA function and dendritic development in pyramidal neurons, as well as abnormalities in sensorimotor gating, albeit without profound memory deficits. We show for the first time that DISC1 is specifically involved in regulating cell surface expression of α2 subunit-containing GABAA receptors in immature developing neurons, but not after full maturation. Notably, pharmacological intervention with α2/3 subtype-selective GABAA receptor positive allosteric modulators during the early postnatal period ameliorates dendritic deficits and behavioral abnormalities induced by knockdown of DISC1. These findings highlight a critical role of DISC1-mediated disruption of postnatal GABA signaling in aberrant PFC maturation and function.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Animales , Modelos Animales de Enfermedad , Electroporación , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Corteza Prefrontal/metabolismo , Subunidades de Proteína , Células Piramidales/metabolismo , Filtrado Sensorial/genética , Filtrado Sensorial/fisiología
5.
J Pharmacol Exp Ther ; 357(2): 264-72, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26945087

RESUMEN

The brain histamine system has been implicated in regulation of sensorimotor gating deficits and in Gilles de la Tourette syndrome. Histamine also regulates alcohol reward and consumption via H3 receptor (H3R), possibly through an interaction with the brain dopaminergic system. Here, we identified the histaminergic mechanism of sensorimotor gating and the role of histamine H3R in the regulation of dopaminergic signaling. We found that H3R knockout mice displayed impaired prepulse inhibition (PPI), indicating deficiency in sensorimotor gating. Histamine H1 receptor knockout and histidine decarboxylase knockout mice had similar PPI as their controls. Dopaminergic drugs increased PPI of H3R knockout mice to the same level as in control mice, suggesting that changes in dopamine receptors might underlie deficient PPI response when H3R is lacking. Striatal dopamine D1 receptor mRNA level was lower, and D1 and D2 receptor-mediated activation of extracellular signal-regulated kinase 1/2 was absent in the striatum of H3R knockout mice, suggesting that H3R is essential for the dopamine receptor-mediated signaling. In conclusion, these findings demonstrate that H3R is an important regulator of sensorimotor gating, and the lack of H3R significantly modifies striatal dopaminergic signaling. These data support the usefulness of H3R ligands in neuropsychiatric disorders with preattentional deficits and disturbances in dopaminergic signaling.


Asunto(s)
Cuerpo Estriado/fisiología , Neuronas Dopaminérgicas/fisiología , Receptores Histamínicos H3/fisiología , Filtrado Sensorial/fisiología , Transducción de Señal/fisiología , Animales , Dopaminérgicos/farmacología , Histidina Descarboxilasa/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/fisiología , Receptores Histamínicos H3/genética , Reflejo de Sobresalto/efectos de los fármacos , Filtrado Sensorial/genética , Transducción de Señal/genética
6.
Mol Psychiatry ; 20(12): 1499-507, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25623945

RESUMEN

Reduced expression of the Gad1 gene-encoded 67-kDa protein isoform of glutamic acid decarboxylase (GAD67) is a hallmark of schizophrenia. GAD67 downregulation occurs in multiple interneuronal sub-populations, including the parvalbumin-positive (PVALB+) cells. To investigate the role of the PV-positive GABAergic interneurons in behavioral and molecular processes, we knocked down the Gad1 transcript using a microRNA engineered to target specifically Gad1 mRNA under the control of Pvalb bacterial artificial chromosome. Verification of construct expression was performed by immunohistochemistry. Follow-up electrophysiological studies revealed a significant reduction in γ-aminobutyric acid (GABA) release probability without alterations in postsynaptic membrane properties or changes in glutamatergic release probability in the prefrontal cortex pyramidal neurons. Behavioral characterization of our transgenic (Tg) mice uncovered that the Pvalb/Gad1 Tg mice have pronounced sensorimotor gating deficits, increased novelty-seeking and reduced fear extinction. Furthermore, NMDA (N-methyl-d-aspartate) receptor antagonism by ketamine had an opposing dose-dependent effect, suggesting that the differential dosage of ketamine might have divergent effects on behavioral processes. All behavioral studies were validated using a second cohort of animals. Our results suggest that reduction of GABAergic transmission from PVALB+ interneurons primarily impacts behavioral domains related to fear and novelty seeking and that these alterations might be related to the behavioral phenotype observed in schizophrenia.


Asunto(s)
Conducta Animal , Glutamato Descarboxilasa/genética , Interneuronas/metabolismo , Parvalbúminas/metabolismo , Esquizofrenia/genética , Animales , Encéfalo/fisiología , Modelos Animales de Enfermedad , Electrofisiología , Conducta Exploratoria , Miedo , Silenciador del Gen , Ketamina/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Filtrado Sensorial/genética , Transmisión Sináptica
7.
Eur Arch Psychiatry Clin Neurosci ; 266(6): 505-12, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26404636

RESUMEN

Schizophrenia (SZ) is a severe mental disorder affecting about 1 % of the human population. Patients show severe deficits in cognitive processing often characterized by an improper filtering of environmental stimuli. Independent genome-wide association studies confirmed a number of risk variants for SZ including several associated with the gene encoding the transcription factor 4 (TCF4). TCF4 is widely expressed in the central nervous system of mice and humans and seems to be important for brain development. Transgenic mice overexpressing murine Tcf4 (Tcf4tg) in the adult brain display cognitive impairments and sensorimotor gating disturbances. To address the question of whether increased Tcf4 gene dosage may affect cognitive flexibility in an auditory associative task, we tested latent inhibition (LI) in female Tcf4tg mice. LI is a widely accepted translational endophenotype of SZ and results from a maladaptive delay in switching a response to a previously unconditioned stimulus when this becomes conditioned. Using an Audiobox, we pre-exposed Tcf4tg mice and their wild-type littermates to either a 3- or a 12-kHz tone before conditioning them to a 12-kHz tone. Tcf4tg animals pre-exposed to a 12-kHz tone showed significantly delayed conditioning when the previously unconditioned tone became associated with an air puff. These results support findings that associate TCF4 dysfunction with cognitive inflexibility and improper filtering of sensory stimuli observed in SZ patients.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Trastornos del Conocimiento/genética , Síndrome de Adaptación General/genética , Filtrado Sensorial/genética , Estimulación Acústica , Animales , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Femenino , Inhibición Psicológica , Locomoción/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Psicoacústica , Factor de Transcripción 4
8.
Learn Mem ; 21(10): 543-55, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25227249

RESUMEN

The Fragile X syndrome (FXS) is the most frequent form of inherited mental disability and is considered a monogenic cause of autism spectrum disorder. FXS is caused by a triplet expansion that inhibits the expression of the FMR1 gene. The gene product, the Fragile X Mental Retardation Protein (FMRP), regulates mRNA metabolism in brain and nonneuronal cells. During brain development, FMRP controls the expression of key molecules involved in receptor signaling, cytoskeleton remodeling, protein synthesis and, ultimately, spine morphology. Symptoms associated with FXS include neurodevelopmental delay, cognitive impairment, anxiety, hyperactivity, and autistic-like behavior. Twenty years ago the first Fmr1 KO mouse to study FXS was generated, and several years later other key models including the mutant Drosophila melanogaster, dFmr1, have further helped the understanding of the cellular and molecular causes behind this complex syndrome. Here, we review to which extent these biological models are affected by the absence of FMRP, pointing out the similarities with the observed human dysfunction. Additionally, we discuss several potential treatments under study in animal models that are able to partially revert some of the FXS abnormalities.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/fisiopatología , Aprendizaje/fisiología , Animales , Ansiedad/genética , Ritmo Circadiano/genética , Drosophila melanogaster , Humanos , Ratones , Ratones Noqueados , Filtrado Sensorial/genética , Transducción de Señal/genética , Conducta Social
9.
Neurobiol Dis ; 63: 210-21, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24239560

RESUMEN

Schizophrenia is a devastating neurodevelopmental disorder that affects approximately 1% of the population. Reduced expression of the 67-kDa protein isoform of glutamic acid decarboxylase (GAD67) is a hallmark of the disease and is encoded by the GAD1 gene. In schizophrenia, GAD67 downregulation occurs in multiple interneuronal subpopulations, including the cannabinoid receptor type 1 positive (CNR1+) cells, but the functional consequences of these disturbances are not well understood. To investigate the role of the CNR1-positive GABA-ergic interneurons in behavioral and molecular processes, we employed a novel, miRNA-mediated transgenic mouse approach. We silenced the Gad1 transcript using a miRNA engineered to specifically target Gad1 mRNA under the control of Cnr1 bacterial artificial chromosome. Behavioral characterization of our transgenic mice showed elevated and persistent conditioned fear associated with an auditory cue and a significantly altered response to an amphetamine challenge. These deficits could not be attributed to sensory deficits or changes in baseline learning and memory. Furthermore, HPLC analyses revealed that Cnr1/Gad1 mice have enhanced serotonin levels, but not dopamine levels in response to amphetamine. Our findings demonstrate that dysfunction of a small subset of interneurons can have a profound effect on behavior and that the GABA-ergic, monoamine, and cannabinoid systems are functionally interconnected. The results also suggest that understanding the function of various interneuronal subclasses might be essential to develop knowledge-based treatment strategies for various mental disorders including schizophrenia and substance abuse.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/citología , Conducta Exploratoria/fisiología , Interneuronas/metabolismo , Receptor Cannabinoide CB1/metabolismo , Anfetamina/farmacología , Analgésicos/farmacología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Psicológico/fisiología , Ciclohexanoles/farmacología , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Locomoción/efectos de los fármacos , Locomoción/genética , Ratones , Ratones Transgénicos , Mutación/genética , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , ARN Mensajero/metabolismo , Receptor Cannabinoide CB1/genética , Filtrado Sensorial/genética
10.
J Neurosci ; 32(32): 10841-53, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22875919

RESUMEN

Delineation of key molecules that act epigenetically to transduce diverse stressors into established patterns of disease would facilitate the advent of preventive and disease-modifying therapeutics for a host of neurological disorders. Herein, we demonstrate that selective overexpression of the stress protein heme oxygenase-1 (HO-1) in astrocytes of novel GFAP.HMOX1 transgenic mice results in subcortical oxidative stress and mitochondrial damage/autophagy; diminished neuronal reelin content (males); induction of Nurr1 and Pitx3 with attendant suppression of their targeting miRNAs, 145 and 133b; increased tyrosine hydroxylase and α-synuclein expression with downregulation of the targeting miR-7b of the latter; augmented dopamine and serotonin levels in basal ganglia; reduced D1 receptor binding in nucleus accumbens; axodendritic pathology and altered hippocampal cytoarchitectonics; impaired neurovascular coupling; attenuated prepulse inhibition (males); and hyperkinetic behavior. The GFAP.HMOX1 neurophenotype bears resemblances to human schizophrenia and other neurodevelopmental conditions and implicates glial HO-1 as a prime transducer of inimical (endogenous and environmental) influences on the development of monoaminergic circuitry. Containment of the glial HO-1 response to noxious stimuli at strategic points of the life cycle may afford novel opportunities for the effective management of human neurodevelopmental and neurodegenerative conditions.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/patología , Regulación del Desarrollo de la Expresión Génica/genética , Hemo-Oxigenasa 1/metabolismo , Esquizofrenia/genética , Esquizofrenia/patología , Estimulación Acústica , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/ultraestructura , Benzamidas/farmacocinética , Benzazepinas/farmacocinética , Monoaminas Biogénicas/metabolismo , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Dopaminérgicos/farmacocinética , Embrión de Mamíferos , Ensayo de Inmunoadsorción Enzimática , Trastornos Neurológicos de la Marcha/etiología , Trastornos Neurológicos de la Marcha/genética , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemo-Oxigenasa 1/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Inhibición Psicológica , Flujometría por Láser-Doppler , Ratones , Ratones Transgénicos , MicroARNs/genética , MicroARNs/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , ARN Mensajero/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Proteína Reelina , Esquizofrenia/fisiopatología , Filtrado Sensorial/genética , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tritio/farmacocinética , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
11.
Synapse ; 67(6): 265-79, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23345061

RESUMEN

The nucleus accumbens (Acb) contains subpopulations of neurons defined by their receptor content and potential involvement in sensorimotor gating and other behaviors that are dysfunctional in schizophrenia. In Acb neurons, the NMDA NR1 (NR1) subunit is coexpressed not only with the dopamine D1 receptor (D1R), but also with the µ-opioid receptor (µ-OR), which mediates certain behaviors that are adversely impacted by schizophrenia. The NMDA-NR1 subunit has been suggested to play a role in the D1R trafficking and behavioral dysfunctions resulting from systemic administration of apomorphine, a D1R and dopamine D2 receptor agonist that impacts prepulse inhibition to auditory-evoked startle (AS). Together, this evidence suggests that the NMDA receptor may regulate D1R trafficking in Acb neurons, including those expressing µ-OR, in animals exposed to auditory startle and apomorphine. We tested this hypothesis by combining spatial-temporal gene deletion technology, dual labeling immunocytochemistry, and behavioral analysis. Deleting NR1 in Acb neurons prevented the increase in the dendritic density of plasma membrane D1Rs in single D1R and dual (D1R and µ-OR) labeled dendrites in the Acb in response to apomorphine and AS. Deleting NR1 also attenuated the decrease in AS induced by apomorphine. In the absence of apomorphine and startle, deletion of Acb NR1 diminished social interaction, without affecting novel object recognition, or open field activity. These results suggest that NR1 expression in the Acb is essential for apomorphine-induced D1R surface trafficking, as well as auditory startle and social behaviors that are impaired in multiple psychiatric disorders.


Asunto(s)
Apomorfina/farmacología , Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Filtrado Sensorial/genética , Animales , Proteínas Portadoras/genética , Membrana Celular/metabolismo , Dendritas/metabolismo , Eliminación de Gen , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Núcleo Accumbens/citología , Núcleo Accumbens/fisiología , Patrones de Reconocimiento Fisiológico , Transporte de Proteínas/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores Opioides mu/metabolismo , Conducta Social
12.
Mol Psychiatry ; 17(1): 85-98, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20956979

RESUMEN

Dysbindin-1 regulates D2-receptor trafficking and is implicated in schizophrenia and related cognitive abnormalities, but whether this molecular effect mediates the clinical manifestations of the disorder is unknown. We explored in dysbindin-1-deficient mice (dys-/-) (1) schizophrenia-related behaviors, (2) molecular and electrophysiological changes in medial prefrontal cortex (mPFC) and (3) the dependence of these on D2-receptor stimulation. Dysbindin-1 disruption altered dopamine-related behaviors and impaired working memory under challenging/stressful conditions. Dys-/- pyramidal neurons in mPFC layers II/III were hyperexcitable at baseline but hypoexcitable following D2 stimulation. Dys-/- were also respectively more and less sensitive to D2 agonist- and antagonist-induced behavioral effects. Dys-/- had reduced expression of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and CaMKKß in mPFC. Chronic D2 agonist treatment reproduced these changes in protein expression, and some of the dys-/- behavioral effects. These results elucidate dysbindin's modulation of D2-related behavior, cortical activity and mPFC CaMK components, implicating cellular and molecular mechanisms of the association of dysbindin with psychosis.


Asunto(s)
Proteínas Portadoras/genética , Dopamina/metabolismo , Corteza Prefrontal/fisiopatología , Receptores de Dopamina D2/metabolismo , Esquizofrenia/patología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Anfetamina/efectos adversos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , Disbindina , Proteínas Asociadas a la Distrofina , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Hipercinesia/tratamiento farmacológico , Hipercinesia/etiología , Hipercinesia/genética , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/patología , Células Piramidales/efectos de los fármacos , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Esquizofrenia/complicaciones , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/genética , Estrés Psicológico/fisiopatología
13.
Pharmacopsychiatry ; 46(7): 286-91, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24105081

RESUMEN

INTRODUCTION: The schizophrenia susceptibility gene neuregulin 1 (NRG1) confers vulnerability to the neurobehavioural eff ects of cannabinoids differently across sexes. Male but not female Nrg1 heterozygous (HET) mice display facilitation of prepulse inhibition (PPI) to acute Δ9-tetrahydrocannabinol (THC) exposure compared to WT controls. We aim to observe whether repeated administration of THC may overcome the acute insensitivity of female Nrg1 HET mice to THC exposure. METHODS: Female Nrg1 HET mice and WT controls were administered THC daily for 21 days, with PPI and anxiety-related behaviour in the light-dark test (LD) examined on the fi rst and last day of treatment and 21 days after cessation of dosing. RESULTS: Following repeated, but not acute THC exposure, female Nrg1 HET mice displayed THC-induced facilitation of PPI which was not observed in WT mice treated with THC. There were no residual eff ects of THC on PPI in either genotype when assessed 21 days following the final THC dose. An anxiogenic response to THC was evident following repeated, but not acute, administration in the LD test in both genotypes. DISCUSSION: These findings show that the acute insensitivity of female Nrg1 HET mice to THC-induced PPI facilitation may be overcome following repeated THC exposure.


Asunto(s)
Dronabinol/farmacología , Heterocigoto , Neurregulina-1/genética , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/genética , Animales , Ansiedad/genética , Femenino , Genotipo , Ratones
14.
PLoS One ; 18(5): e0277446, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37205689

RESUMEN

Protein Tyrosine Phosphatase receptor type D (PTPRD) is a member of the protein tyrosine phosphatase family that mediates cell adhesion and synaptic specification. Genetic studies have linked Ptprd to several neuropsychiatric phenotypes, including Restless Leg Syndrome (RLS), opioid abuse disorder, and antipsychotic-induced weight gain. Genome-wide association studies (GWAS) of either pediatric obsessive-compulsive traits, or Obsessive-Compulsive Disorder (OCD), have identified loci near PTPRD as genome-wide significant, or strongly suggestive for this trait. We assessed Ptprd wild-type (WT), heterozygous (HT), and knockout (KO) mice for behavioral dimensions that are altered in OCD, including anxiety and exploration (open field test, dig test), perseverative behavior (splash-induced grooming, spatial d), sensorimotor gating (prepulse inhibition), and home cage goal-directed behavior (nest building). No effect of genotype was observed in any measure of the open field test, dig test, or splash test. However, Ptprd KO mice of both sexes showed impairments in nest building behavior. Finally, female, but not male, Ptprd KO mice showed deficits in prepulse inhibition, an operational measure of sensorimotor gating that is reduced in female, but not male, OCD patients. Our results indicate that constitutive lack of Ptprd may contribute to the development of certain domains that are altered OCD, including goal-directed behavior, and reduced sensorimotor gating specifically in females.


Asunto(s)
Estudio de Asociación del Genoma Completo , Trastorno Obsesivo Compulsivo , Masculino , Femenino , Animales , Ratones , Objetivos , Trastorno Obsesivo Compulsivo/genética , Genotipo , Inhibición Prepulso , Ratones Noqueados , Filtrado Sensorial/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética
15.
J Neurosci ; 31(18): 6684-91, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21543597

RESUMEN

In a large-scale meta-analysis, it has been recently shown that the transcription factor 4 (TCF4) gene is among the most prominent susceptibility genes for schizophrenia. Moreover, transgenic mice overexpressing TCF4 in the brain display a reduction of sensorimotor gating measured by prepulse inhibition (PPI) of the acoustic startle response (ASR). PPI is heritable and has been established as an important translational endophenotype of schizophrenia. We therefore investigated the impact of the schizophrenia susceptibility gene TCF4 (rs9960767) on sensorimotor gating of the ASR in healthy humans and in patients with a schizophrenia spectrum disorder. We assessed PPI, startle reactivity, and habituation of the ASR in two independent samples. The first sample consisted of 107 healthy volunteers from London, UK. The second sample was a schizophrenia spectrum group (n = 113) of 73 schizophrenia patients and 40 individuals at high risk for schizophrenia from Bonn, Germany (total sample n = 220). In both samples, PPI was strongly decreased in carriers of the schizophrenia risk allele C of the TCF4 gene (meta-analysis across both samples: p = 0.00002), whereas startle reactivity and habituation were unaffected by TCF4 genotype. Sensorimotor gating is modulated by TCF4 genotype, indicating an influential role of TCF4 gene variations in the development of early information-processing deficits in schizophrenia.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Polimorfismo Genético/genética , Reflejo de Sobresalto/genética , Esquizofrenia/genética , Filtrado Sensorial/genética , Factores de Transcripción/genética , Estimulación Acústica , Adolescente , Adulto , Alelos , Análisis de Varianza , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Factor de Transcripción 4
16.
Dev Neurosci ; 34(2-3): 159-73, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22571986

RESUMEN

Mice with knockdown of the N-methyl-D-aspartate (NMDA) receptor NR1 subunit, encoded by the gene Grin1, have been investigated as a model for the intrinsic NMDA hypofunction hypothesized for schizophrenia. Previous work has shown that adult Grin1 mutant mice have overt deficits in habituation and sensorimotor gating, exaggerated reactivity to environmental stimuli, reduced social approach, and other alterations that reflect behavioral manifestations of schizophrenia. In humans, the emergence of overt symptoms of the disorder typically occurs in adolescence or early adulthood, suggesting a role for aberrant maturation of NMDA receptor signaling in symptom onset. The following study evaluated Grin1 mutant mice for abnormal behavioral phenotypes during the preweaning, adolescent, and adult periods. Measures included open field activity, prepulse inhibition of acoustic startle responses, and social preference in a three-chamber choice task. Mice from the C57BL/6J inbred strain, one of the parental strains for the Grin1 line, were also tested. The results showed that developmental reduction of NMDA receptor function led to significant alterations in behavior during the second and third weeks of life, including exaggerated startle responses and sensorimotor gating deficits on postnatal day 13, and pronounced hypersociability in adolescence. Male Grin1 mutant mice were more susceptible than female mice to the detrimental effects of decreased NMDA signaling. Overall, these findings provide evidence that reduced Grin1 function leads to abnormal phenotypes in the preweaning period, and that deficient NMDA signaling can lead to both overt hypersociability or marked asociality, dependent upon sex and age.


Asunto(s)
Conducta Animal/fisiología , Receptores de N-Metil-D-Aspartato/genética , Reflejo de Sobresalto/genética , Esquizofrenia/genética , Filtrado Sensorial/genética , Conducta Social , Factores de Edad , Animales , Conducta de Elección/fisiología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/fisiopatología , Factores Sexuales
17.
Int J Neuropsychopharmacol ; 15(10): 1427-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22244514

RESUMEN

Converging evidence from pharmacological investigations, genetic association studies and schizophrenia research indicates an important influence of the dopamine system on sensorimotor gating as measured by prepulse inhibition (PPI) of the acoustic startle response. In particular, D2 receptor agonists have been shown to disrupt PPI in humans and rodents. In the present study, we investigated the associations of two functional DRD2 related single nucleotide polymorphisms (rs4648317 and rs1800497, the latter also known as DRD2/ANKK1 Taq1A) with PPI in two independent healthy human samples (overall n=197; Munich n=101; London n=96). Taq1A is a prominent marker of striatal D2 receptor signalling and was therefore hypothesized to impact on PPI. In line with our hypothesis, we report here reduced PPI levels in individuals with higher striatal D2 receptor signalling as indicated by the Taq1A genotype. Meta-analysis across both samples confirmed this finding. In contrast, an association between rs4648317 and PPI found in the Munich sample could not be confirmed in the London sample. Overall, the present study helps to bridge the gap between pharmacological manipulations of PPI and molecular genetics of the dopaminergic system.


Asunto(s)
Estimulación Acústica/métodos , Genotipo , Polimorfismo de Nucleótido Simple/genética , Receptores de Dopamina D2/genética , Filtrado Sensorial/genética , Transducción de Señal/genética , Adulto , Femenino , Humanos , Masculino , Adulto Joven
18.
Int J Neuropsychopharmacol ; 15(8): 1073-86, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21777509

RESUMEN

Brain-derived neurotrophic factor (BDNF) signalling through its receptor, TrkB is known to regulate GABAergic function and glutamic acid decarboxylase (GAD) 67 expression in neurons. Alterations in BDNF signalling have been implicated in the pathophysiology of schizophrenia and as a result, they are a potential therapeutic target. Interestingly, heterozygous reeler mice (HRM) have decreased GAD67 expression in the frontal cortex and hippocampus and they exhibit many behavioural and neurochemical abnormalities similar to schizophrenia. In this study, we evaluated the potential of cysteamine, a neuroprotective compound to improve the deficits in GAD67 expression and cognitive function in HRM. We found that cysteamine administration (150 mg/kg.d, through drinking water) for 30 d significantly ameliorated the decreases in GAD67, mature BDNF and full-length TrkB protein levels found in frontal cortex and hippocampus of HRM. A significant attenuation of the increased levels of truncated BDNF in frontal cortex and hippocampus, as well as truncated TrkB in frontal cortex of HRM was also observed following cysteamine treatment. In behavioural studies, HRM were impaired in a Y-maze spatial recognition memory task, but not in a spontaneous alternation task or a sensorimotor, prepulse inhibition (PPI) procedure. Cysteamine improved Y-maze spatial recognition in HRM to the level of wide-type controls and it improved PPI in both wild-type and HRM. Finally, mice deficient in TrkB, showed a reduced response to cysteamine in GAD67 expression suggesting that TrkB signalling plays an important role in GAD67 regulation by cysteamine.


Asunto(s)
Cisteamina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/metabolismo , Memoria/efectos de los fármacos , Percepción Espacial/efectos de los fármacos , Estimulación Acústica/efectos adversos , Análisis de Varianza , Animales , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Regulación de la Expresión Génica/genética , Glutamato Descarboxilasa/genética , Heterocigoto , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Noqueados , Ratones Mutantes Neurológicos , Receptor trkB/deficiencia , Receptor trkB/metabolismo , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/genética , Factores de Tiempo
19.
Encephale ; 38 Suppl 3: S103-9, 2012 Dec.
Artículo en Francés | MEDLINE | ID: mdl-23279983

RESUMEN

It is proposed an historical approach to concepts leading to the development of operational paradigms for measuring objectives neurophysiological endophenotypes. It is hypothesized that psychiatric interest for paradigms measuring Event-Related Potential (ERP) come from Bleuler (1911) and McGhie and Chapman (1961) phenomenological and clinical descriptions. They noted, first that patients with schizophrenia generally feel as if they are being flooded by an overwhelming mass of sensory input combined with a heightened sensory perception, second that they were distractible to irrelevant sensory stimuli. These subjective abnormalities may be related, first to inability to filter incongruent information measured in a double click paradigm by a deficit in P50 amplitude gating, and second to an inability to select a stimulus of interest measured in the oddball paradigm by a deficit in P300 amplitude. The analysis of these P50 and P300 ERP in cohorts of patients with schizophrenia found most of Gottesman endophenotype criteria. P50 and P300 ERP are therefore relevant neurophysiological endophenotypes. However, from a clinical point of view, these endophenotypes lack specificity. The hypothesis of this article leads us to formulate ways of research. It is shown the value of combining objective neurophysiological measures with subjective measures using self-administered questionnaires ("offline") or psychophysiological tests ("online") to develop rigorous neurophysiological experimental paradigms especially as clinical observations of their origins are not forgotten.


Asunto(s)
Nivel de Alerta/genética , Nivel de Alerta/fisiología , Encéfalo/fisiopatología , Endofenotipos , Potenciales Evocados/genética , Potenciales Evocados/fisiología , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Psicología del Esquizofrénico , Filtrado Sensorial/genética , Filtrado Sensorial/fisiología , Estimulación Acústica , Atención/fisiología , Potenciales Relacionados con Evento P300/genética , Potenciales Relacionados con Evento P300/fisiología , Predisposición Genética a la Enfermedad/genética , Humanos , Esquizofrenia/diagnóstico
20.
J Neurosci ; 30(2): 556-67, 2010 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20071518

RESUMEN

The membrane protein Nogo-A, which is predominantly expressed by oligodendrocytes in the adult CNS and by neurons mainly during development, is well known for limiting neurite outgrowth and regeneration in the injured mammalian CNS. In addition, it has recently been proposed that abnormal Nogo-A expression or Nogo receptor (NgR) mutations may confer genetic risks for neuropsychiatric disorders of presumed neurodevelopmental origin, such as schizophrenia. We therefore evaluated whether Nogo-A deletion may lead to schizophrenia-like abnormalities in a mouse model of genetic Nogo-A deficiency. Here, we show that systemic, lifelong knock-out of the Nogo-A gene can lead to specific behavioral abnormalities resembling schizophrenia-related endophenotypes: deficient sensorimotor gating, disrupted latent inhibition, perseverative behavior, and increased sensitivity to the locomotor stimulating effects of amphetamine. These behavioral phenotypes were accompanied by altered monoaminergic transmitter levels in specific striatal and limbic structures, as well as changes in dopamine D2 receptor expression in the same brain regions. Nogo-A deletion was further associated with elevated expression of growth-related markers. In contrast, acute antibody-mediated Nogo-A neutralization in adult wild-type mice failed to produce such phenotypes, suggesting that the phenotypes observed in the knock-out mice might be of developmental origin, and that Nogo-A normally subserves critical functions in neurodevelopment. This study provides the first experimental demonstration that Nogo-A bears neuropsychiatric relevance, and alterations in its expression may be one etiological factor in schizophrenia and related disorders.


Asunto(s)
Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/fisiología , Fenotipo , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Eliminación de Secuencia/genética , Estimulación Acústica/efectos adversos , Anfetamina/farmacología , Análisis de Varianza , Animales , Anticuerpos/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Estimulantes del Sistema Nervioso Central/farmacología , Cromatografía Líquida de Alta Presión/métodos , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Inhibición Psicológica , Relaciones Interpersonales , Aprendizaje/fisiología , Locomoción/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de la Mielina/inmunología , Neurotransmisores/metabolismo , Proteínas Nogo , Corteza Prefrontal/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/patología , Filtrado Sensorial/genética , Filtrado Sensorial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA