Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Transl Psychiatry ; 11(1): 182, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753722

RESUMEN

Low-frequency 1q21.1 distal deletion and duplication copy number variant (CNV) carriers are predisposed to multiple neurodevelopmental disorders, including schizophrenia, autism and intellectual disability. Human carriers display a high prevalence of micro- and macrocephaly in deletion and duplication carriers, respectively. The underlying brain structural diversity remains largely unknown. We systematically called CNVs in 38 cohorts from the large-scale ENIGMA-CNV collaboration and the UK Biobank and identified 28 1q21.1 distal deletion and 22 duplication carriers and 37,088 non-carriers (48% male) derived from 15 distinct magnetic resonance imaging scanner sites. With standardized methods, we compared subcortical and cortical brain measures (all) and cognitive performance (UK Biobank only) between carrier groups also testing for mediation of brain structure on cognition. We identified positive dosage effects of copy number on intracranial volume (ICV) and total cortical surface area, with the largest effects in frontal and cingulate cortices, and negative dosage effects on caudate and hippocampal volumes. The carriers displayed distinct cognitive deficit profiles in cognitive tasks from the UK Biobank with intermediate decreases in duplication carriers and somewhat larger in deletion carriers-the latter potentially mediated by ICV or cortical surface area. These results shed light on pathobiological mechanisms of neurodevelopmental disorders, by demonstrating gene dose effect on specific brain structures and effect on cognitive function.


Asunto(s)
Variaciones en el Número de Copia de ADN , Esquizofrenia , Encéfalo/diagnóstico por imagen , Deleción Cromosómica , Cognición , Femenino , Humanos , Masculino , Esquizofrenia/genética
2.
Transl Psychiatry ; 10(1): 239, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32681022

RESUMEN

The schizophrenia-associated gene, BRD1, encodes an epigenetic regulator in which chromatin interactome is enriched with genes implicated in mental health. Alterations in histone modifications and epigenetic regulation contribute to brain transcriptomic changes in affective disorders and preclinical data supports a role for BRD1 in psychopathology. However, the implication of BRD1 on affective pathology remains poorly understood. In this study, we assess affective behaviors and associated neurobiology in Brd1+/- mice along with their responses to Fluoxetine and Imipramine. This involves behavioral, neurostructural, and neurochemical characterizations along with regional cerebral gene expression profiling combined with integrative functional genomic analyses. We report behavioral changes in female Brd1+/- mice with translational value to depressive symptomatology that can be alleviated by the administration of antidepressant medications. Behavioral changes are accompanied by altered brain morphometry and imbalances in monoaminergic systems. In accordance, gene expression changes across brain tissues reveal altered neurotransmitter signaling and cluster in functional pathways associated with depression including 'Adrenergic-, GPCR-, cAMP-, and CREB/CREM-signaling'. Integrative gene expression analysis specifically links changes in amygdaloid intracellular signaling activity to the behavioral treatment response in Brd1+/- mice. Collectively, our study highlights the importance of BRD1 as a modulator of affective pathology and adds to our understanding of the molecular mechanisms underlying affective disorders and their treatment response.


Asunto(s)
Histona Acetiltransferasas , Esquizofrenia , Animales , Depresión/genética , Epigénesis Genética , Femenino , Expresión Génica , Ratones , Esquizofrenia/genética
3.
Transl Psychiatry ; 8(1): 247, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30429456

RESUMEN

The 22q11.2 deletion syndrome (22q11.2DS) confers high risk of neurodevelopmental disorders such as schizophrenia and attention-deficit hyperactivity disorder. These disorders are associated with attentional impairment, the remediation of which is important for successful therapeutic intervention. We assessed a 22q11.2DS mouse model (Df(h22q11)/+) on a touchscreen rodent continuous performance test (rCPT) of attention and executive function that is analogous to human CPT procedures. Relative to wild-type littermates, Df(h22q11)/+ male mice showed impaired attentional performance as shown by decreased correct response ratio (hit rate) and a reduced ability to discriminate target stimuli from non-target stimuli (discrimination sensitivity, or d'). The Df(h22q11)/+ model exhibited decreased prefrontal cortical-hippocampal oscillatory synchrony within multiple frequency ranges during quiet wakefulness, which may represent a biomarker of cognitive dysfunction. The stimulant amphetamine (0-1.0 mg/kg, i.p.) dose-dependently improved d' in Df(h22q11)/+ mice whereas the highest dose of modafinil (40 mg/kg, i.p.) exacerbated their d' impairment. This is the first report to directly implicate attentional impairment in a 22q11.2DS mouse model, mirroring a key endophenotype of the human disorder. The capacity of the rCPT to detect performance impairments in the 22q11.2DS mouse model, and improvement following psychostimulant-treatment, highlights the utility and translational potential of the Df(h22q11)/+ model and this automated behavioral procedure.


Asunto(s)
Atención/fisiología , Conducta Animal/fisiología , Estimulantes del Sistema Nervioso Central/farmacología , Disfunción Cognitiva/fisiopatología , Síndrome de DiGeorge/fisiopatología , Sincronización de Fase en Electroencefalografía/fisiología , Función Ejecutiva/fisiología , Hipocampo/fisiopatología , Corteza Prefrontal/fisiopatología , Desempeño Psicomotor/fisiología , Anfetamina/farmacología , Animales , Atención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/administración & dosificación , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad , Función Ejecutiva/efectos de los fármacos , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Modafinilo/farmacología , Corteza Prefrontal/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos
4.
Transl Psychiatry ; 7(11): 1261, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29187755

RESUMEN

1q21.1 hemizygous microdeletion is a copy number variant leading to eightfold increased risk of schizophrenia. In order to investigate biological alterations induced by this microdeletion, we generated a novel mouse model (Df(h1q21)/+) and characterized it in a broad test battery focusing on schizophrenia-related assays. Df(h1q21)/+ mice displayed increased hyperactivity in response to amphetamine challenge and increased sensitivity to the disruptive effects of amphetamine and phencyclidine hydrochloride (PCP) on prepulse inhibition. Probing of the direct dopamine (DA) pathway using the DA D1 receptor agonist SKF-81297 revealed no differences in induced locomotor activity compared to wild-type mice, but Df(h1q21)/+ mice showed increased sensitivity to the DA D2 receptor agonist quinpirole and the D1/D2 agonist apomorphine. Electrophysiological characterization of DA neuron firing in the ventral tegmental area revealed more spontaneously active DA neurons and increased firing variability in Df(h1q21)/+ mice, and decreased feedback reduction of DA neuron firing in response to amphetamine. In a range of other assays, Df(h1q21)/+ mice showed no difference from wild-type mice: gross brain morphology and basic functions such as reflexes, ASR, thermal pain sensitivity, and motor performance were unaltered. Similarly, anxiety related measures, baseline prepulse inhibition, and seizure threshold were unaltered. In addition to the central nervous system-related phenotypes, Df(h1q21)/+ mice exhibited reduced head-to tail length, which is reminiscent of the short stature reported in humans with 1q21.1 deletion. With aspects of both construct and face validity, the Df(h1q21)/+ model may be used to gain insight into schizophrenia-relevant alterations in dopaminergic transmission.


Asunto(s)
Anomalías Múltiples , Conducta Animal , Deleción Cromosómica , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Neuronas Dopaminérgicas/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Megalencefalia , Núcleo Accumbens/metabolismo , Inhibición Prepulso , Receptores Dopaminérgicos/metabolismo , Esquizofrenia , Área Tegmental Ventral/metabolismo , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Anfetamina/farmacología , Animales , Apomorfina/farmacología , Conducta Animal/efectos de los fármacos , Benzazepinas/farmacología , Cromosomas Humanos Par 1/metabolismo , Modelos Animales de Enfermedad , Agonistas de Dopamina/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Megalencefalia/metabolismo , Megalencefalia/patología , Megalencefalia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Fenciclidina/farmacología , Fenotipo , Inhibición Prepulso/efectos de los fármacos , Quinpirol/farmacología , Receptores Dopaminérgicos/efectos de los fármacos , Esquizofrenia/metabolismo , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Área Tegmental Ventral/efectos de los fármacos
5.
J Psychiatry Neurosci ; 42(1): 48-58, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27391101

RESUMEN

BACKGROUND: The hemizygous 22q11.2 microdeletion is a common copy number variant in humans. The deletion confers high risk for neurodevelopmental disorders, including autism and schizophrenia. Up to 41% of deletion carriers experience psychotic symptoms. METHODS: We present a new mouse model (Df(h22q11)/+) of the deletion syndrome (22q11.2DS) and report on, to our knowledge, the most comprehensive study undertaken to date in 22q11.2DS models. The study was conducted in male mice. RESULTS: We found elevated postpubertal N-methyl-D-aspartate (NMDA) receptor antagonist-induced hyperlocomotion, age-independent prepulse inhibition (PPI) deficits and increased acoustic startle response (ASR). The PPI deficit and increased ASR were resistant to antipsychotic treatment. The PPI deficit was not a consequence of impaired hearing measured by auditory brain stem responses. The Df(h22q11)/+ mice also displayed increased amplitude of loudness-dependent auditory evoked potentials. Prefrontal cortex and dorsal striatal elevations of the dopamine metabolite DOPAC and increased dorsal striatal expression of the AMPA receptor subunit GluR1 was found. The Df(h22q11)/+ mice did not deviate from wild-type mice in a wide range of other behavioural and biochemical assays. LIMITATIONS: The 22q11.2 microdeletion has incomplete penetrance in humans, and the severity of disease depends on the complete genetic makeup in concert with environmental factors. In order to obtain more marked phenotypes reflecting the severe conditions related to 22q11.2DS it is suggested to expose the Df(h22q11)/+ mice to environmental stressors that may unmask latent psychopathology. CONCLUSION: The Df(h22q11)/+ model will be a valuable tool for increasing our understanding of the etiology of schizophrenia and other psychiatric disorders associated with the 22q11DS.


Asunto(s)
Envejecimiento/fisiología , Síndrome de DiGeorge/fisiopatología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Filtrado Sensorial/fisiología , Ácido 3,4-Dihidroxifenilacético/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Percepción Auditiva/fisiología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reflejo de Sobresalto/fisiología
6.
Biol Psychiatry ; 82(1): 62-76, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27837920

RESUMEN

BACKGROUND: The schizophrenia-associated BRD1 gene encodes a transcriptional regulator whose comprehensive chromatin interactome is enriched with schizophrenia risk genes. However, the biology underlying the disease association of BRD1 remains speculative. METHODS: This study assessed the transcriptional drive of a schizophrenia-associated BRD1 risk variant in vitro. Accordingly, to examine the effects of reduced Brd1 expression, we generated a genetically modified Brd1+/- mouse and subjected it to behavioral, electrophysiological, molecular, and integrative genomic analyses with focus on schizophrenia-relevant parameters. RESULTS: Brd1+/- mice displayed cerebral histone H3K14 hypoacetylation and a broad range of behavioral changes with translational relevance to schizophrenia. These behaviors were accompanied by striatal dopamine/serotonin abnormalities and cortical excitation-inhibition imbalances involving loss of parvalbumin immunoreactive interneurons. RNA-sequencing analyses of cortical and striatal micropunches from Brd1+/- and wild-type mice revealed differential expression of genes enriched for schizophrenia risk, including several schizophrenia genome-wide association study risk genes (e.g., calcium channel subunits [Cacna1c and Cacnb2], cholinergic muscarinic receptor 4 [Chrm4)], dopamine receptor D2 [Drd2], and transcription factor 4 [Tcf4]). Integrative analyses further found differentially expressed genes to cluster in functional networks and canonical pathways associated with mental illness and molecular signaling processes (e.g., glutamatergic, monoaminergic, calcium, cyclic adenosine monophosphate [cAMP], dopamine- and cAMP-regulated neuronal phosphoprotein 32 kDa [DARPP-32], and cAMP responsive element binding protein signaling [CREB]). CONCLUSIONS: Our study bridges the gap between genetic association and pathogenic effects and yields novel insights into the unfolding molecular changes in the brain of a new schizophrenia model that incorporates genetic risk at three levels: allelic, chromatin interactomic, and brain transcriptomic.


Asunto(s)
Conducta Animal/fisiología , Expresión Génica/genética , Histona Acetiltransferasas/fisiología , Esquizofrenia/genética , Transmisión Sináptica/genética , Acetilación , Animales , Animales Modificados Genéticamente/genética , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Histona Acetiltransferasas/genética , Histonas/metabolismo , Interneuronas/fisiología , Ratones , Serotonina/metabolismo
7.
Cereb Cortex ; 26(10): 3991-4003, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27507786

RESUMEN

A chromosomal microdeletion at the 22q11.2 locus is associated with extensive cognitive impairments, schizophrenia and other psychopathology in humans. Previous reports indicate that mouse models of the 22q11.2 microdeletion syndrome (22q11.2DS) may model the genetic basis of cognitive deficits relevant for neuropsychiatric disorders such as schizophrenia. To assess the models usefulness for drug discovery, a novel mouse (Df(h22q11)/+) was assessed in an extensive battery of cognitive assays by partners within the NEWMEDS collaboration (Innovative Medicines Initiative Grant Agreement No. 115008). This battery included classic and touchscreen-based paradigms with recognized sensitivity and multiple attempts at reproducing previously published findings in 22q11.2DS mouse models. This work represents one of the most comprehensive reports of cognitive functioning in a transgenic animal model. In accordance with previous reports, there were non-significant trends or marginal impairment in some tasks. However, the Df(h22q11)/+ mouse did not show comprehensive deficits; no robust impairment was observed following more than 17 experiments and 14 behavioral paradigms. Thus - within the current protocols - the 22q11.2DS mouse model fails to mimic the cognitive alterations observed in human 22q11.2 deletion carriers. We suggest that the 22q11.2DS model may induce liability for cognitive dysfunction with additional "hits" being required for phenotypic expression.


Asunto(s)
Cognición , Síndrome de DiGeorge/psicología , Modelos Animales de Enfermedad , Ratones Transgénicos , Animales , Atención , Estudios de Cohortes , Discriminación en Psicología , Descubrimiento de Drogas , Función Ejecutiva , Femenino , Inhibición Psicológica , Aprendizaje , Masculino , Memoria , Ratones Endogámicos C57BL , Pruebas Neuropsicológicas , Fenotipo , Investigación Biomédica Traslacional
8.
Psychopharmacology (Berl) ; 233(11): 2151-2163, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26983414

RESUMEN

RATIONALE: A microdeletion at locus 15q13.3 is associated with high incidence rates of psychopathology, including schizophrenia. A mouse model of the 15q13.3 microdeletion syndrome has been generated (Df[h15q13]/+) with translational utility for modelling schizophrenia-like pathology. Among other deficits, schizophrenia is characterised by dysfunctions in prefrontal cortical (PFC) inhibitory circuitry and attention. OBJECTIVES: The objective of this study is to assess PFC-dependent functioning in the Df(h15q13)/+ mouse using electrophysiological, pharmacological, and behavioural assays. METHOD: Experiments 1-2 investigated baseline firing and auditory-evoked responses of PFC interneurons and pyramidal neurons. Experiment 3 measured pyramidal firing in response to intra-PFC GABAA receptor antagonism. Experiments 4-6 assessed PFC-dependent attentional functioning through the touchscreen 5-choice serial reaction time task (5-CSRTT). Experiments 7-12 assessed reversal learning, paired-associate learning, extinction learning, progressive ratio, trial-unique non-match to sample, and object recognition. RESULTS: In experiments 1-3, the Df(h15q13)/+ mouse showed reduced baseline firing rate of fast-spiking interneurons and in the ability of the GABAA receptor antagonist gabazine to increase the firing rate of pyramidal neurons. In assays of auditory-evoked responses, PFC interneurons in the Df(h15q13)/+ mouse had reduced detection amplitudes and increased detection latencies, while pyramidal neurons showed increased detection latencies. In experiments 4-6, the Df(h15q13)/+ mouse showed a stimulus duration-dependent decrease in percent accuracy in the 5-CSRTT. The impairment was insensitive to treatment with the partial α7nAChR agonist EVP-6124. The Df(h15q13)/+ mouse showed no cognitive impairments in experiments 7-12. CONCLUSION: The Df(h15q13)/+ mouse has multiple dysfunctions converging on disrupted PFC processing as measured by several independent assays of inhibitory transmission and attentional function.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Eliminación de Gen , Corteza Prefrontal/fisiopatología , Esquizofrenia/genética , Esquizofrenia/fisiopatología , Psicología del Esquizofrénico , Animales , Trastorno por Déficit de Atención con Hiperactividad/psicología , Conducta Animal/efectos de los fármacos , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 15/genética , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos/efectos de los fármacos , Extinción Psicológica/efectos de los fármacos , Antagonistas del GABA/farmacología , Humanos , Discapacidad Intelectual/genética , Interneuronas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Piramidales/efectos de los fármacos , Piridazinas/farmacología , Tiempo de Reacción/efectos de los fármacos , Receptores de GABA-A/efectos de los fármacos , Aprendizaje Inverso/efectos de los fármacos , Convulsiones/genética
9.
Biol Psychiatry ; 76(2): 128-37, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24090792

RESUMEN

BACKGROUND: Genome-wide scans have uncovered rare copy number variants conferring high risk of psychiatric disorders. The 15q13.3 microdeletion is associated with a considerably increased risk of idiopathic generalized epilepsy, intellectual disability, and schizophrenia. METHODS: A 15q13.3 microdeletion mouse model (Df[h15q13]/+) was generated by hemizygous deletion of the orthologous region and characterized with focus on schizophrenia- and epilepsy-relevant parameters. RESULTS: Df(h15q13)/+ mice showed marked changes in neuronal excitability in acute seizure assays, with increased propensity to develop myoclonic and absence-like seizures but decreased propensity for clonic and tonic seizures. Furthermore, they had impaired long-term spatial reference memory and a decreased theta frequency in hippocampus and prefrontal cortex. Electroencephalogram characterization revealed auditory processing deficits similar to those observed in schizophrenia. Gamma band power was increased during active state, but evoked gamma power following auditory stimulus (40 Hz) was dramatically reduced, mirroring observations in patients with schizophrenia. In addition, Df(h15q13)/+ mice showed schizophrenia-like decreases in amplitudes of auditory evoked potentials. Although displaying a grossly normal behavior, Df(h15q13)/+ mice are more aggressive following exposure to mild stressors, similar to what is described in human deletion carriers. Furthermore, Df(h15q13)/+ mice have increased body weight, and a similar increase in body weight was subsequently found in a sample of human subjects with 15q13.3 deletion. CONCLUSIONS: The Df(h15q13)/+ mouse shows similarities to several alterations related to the 15q13.3 microdeletion syndrome, epilepsy, and schizophrenia, offering a novel tool for addressing the underlying biology of these diseases.


Asunto(s)
Encéfalo/fisiopatología , Trastornos de los Cromosomas/genética , Modelos Animales de Enfermedad , Epilepsia/genética , Discapacidad Intelectual/genética , Ratones , Esquizofrenia/genética , Convulsiones/genética , Animales , Conducta Animal/fisiología , Peso Corporal/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Electroencefalografía , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad
10.
Neuropharmacology ; 72: 157-68, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23643744

RESUMEN

The underlying mechanism of the GABAergic deficits observed in schizophrenia has been proposed to involve NMDA receptor hypofunction. An emerging treatment strategy therefore aims at enhancing GABAergic signalling by increasing the excitatory transmission onto interneurons. We wanted to determine whether behavioural and GABAergic functional deficits induced by the NMDA receptor channel blocker, phencyclidine (PCP), could be reversed by repeated administration of two drugs known to enhance GABAergic transmission: the positive allosteric modulator (PAM) of the metabotropic glutamate receptor 5 (mGluR5), ADX47273, and the partial agonist of the α7 nicotinic acetylcholine receptor (α7 nAChR), SSR180711. Adolescent rats (4-5 weeks) subjected to PCP treatment during the second postnatal week displayed a consistent deficit in prepulse inhibition (PPI), which was reversed by a one-week treatment with ADX47273 or SSR180711. We examined GABAergic transmission by whole cell patch-clamp recordings of miniature inhibitory postsynaptic currents (mIPSC) in pyramidal neurons in layer II/III of prefrontal cortex (PFC) and by activation of extrasynaptic δ-containing GABAA receptors by THIP. Following PCP treatment, pyramidal neurons displayed a reduced mIPSC frequency and up-regulation of extrasynaptic THIP-induced current. ADX47273 treatment restored this up-regulation of THIP-induced current. Reduced receptor function seems to be the underlying cause of the reported changes, since repeated treatment with ADX47273 and SSR180711 decreased the induction of spontaneous inhibitory current caused by acute and direct agonism of mGluR5s and α7 nAChRs in slices. These results show that repeated administration of ADX47273 or SSR180711 reverses certain behavioural and functional deficits induced by PCP, likely through down-regulation or desensitisation of mGluR5s and α7 nAChRs, respectively.


Asunto(s)
Neuronas GABAérgicas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Receptor del Glutamato Metabotropico 5/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Colinérgicos/farmacología , Antagonistas de Aminoácidos Excitadores/efectos adversos , Femenino , Lóbulo Frontal/citología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Oxadiazoles/metabolismo , Oxadiazoles/farmacología , Fenciclidina/efectos adversos , Piperidinas/metabolismo , Piperidinas/farmacología , Embarazo , Ratas , Factores de Tiempo
11.
BMC Med ; 9: 40, 2011 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-21501494

RESUMEN

BACKGROUND: Accumulating evidence suggests that the brain's nitric oxide (NO) signalling system may be involved in the pathophysiology of schizophrenia and could thus constitute a novel treatment target. The study was designed to investigate the benefit of L-lysine, an amino acid that interferes with NO production, as an add-on treatment for schizophrenia. METHODS: L-lysine, 6 g/day, was administered to 10 patients with schizophrenia as an adjunctive to their conventional antipsychotic medication. The study was designed as a single-blinded, cross-over study where patients were randomly assigned to initial treatment with either L-lysine or placebo and screened at baseline, after four weeks when treatment was crossed over, and after eight weeks. RESULTS: L-lysine treatment caused a significant increase in blood concentration of L-lysine and was well tolerated. A significant decrease in positive symptom severity, measured by the Positive And Negative Syndrome Scale (PANSS), was detected. A certain decrease in score was also observed during placebo treatment and the effects on PANSS could not unequivocally be assigned to the L-lysine treatment. Furthermore, performance on the Wisconsin Card Sorting Test was significantly improved compared to baseline, an effect probably biased by training. Subjective reports from three of the patients indicated decreased symptom severity and enhanced cognitive functioning. CONCLUSIONS: Four-week L-lysine treatment of 6 g/day caused a significant increase in blood concentration of L-lysine that was well tolerated. Patients showed a significant decrease in positive symptoms as assessed by PANSS in addition to self-reported symptom improvement by three patients. The NO-signalling pathway is an interesting, potentially new treatment target for schizophrenia; however, the effects of L-lysine need further evaluation to decide the amino acid's potentially beneficial effects on symptom severity in schizophrenia.


Asunto(s)
Antipsicóticos/administración & dosificación , Lisina/administración & dosificación , Esquizofrenia/tratamiento farmacológico , Adulto , Antipsicóticos/efectos adversos , Estudios Cruzados , Quimioterapia Combinada/efectos adversos , Quimioterapia Combinada/métodos , Femenino , Humanos , Lisina/efectos adversos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Placebos/administración & dosificación , Resultado del Tratamiento
12.
Eur Neuropsychopharmacol ; 21(3): 248-53, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21111580

RESUMEN

A well established theory proposes that glutamate signalling via the NMDA receptor is compromised in patients with schizophrenia. Deficits related to NMDA receptor signalling can be observed in several brain regions including the prefrontal cortex (PFC), an area extensively linked to the cognitive dysfunction in this disease and notably affected by NMDA receptor antagonists such as phencyclidine (PCP). In addition, a number of studies suggest that normalizing of PFC function could constitute a treatment rationale for schizophrenia. To further study the role of PFC function we investigated the effect of local PFC NMDA receptor blockade on impaired prepulse inhibition (PPI) induced by systemic administration of PCP. Mice received prefrontal injections of PCP (0.01, 0.1 or 1mM) before PCP treatment (5mg/kg) and were thereafter tested for PPI. PCP induced deficits in PPI were ameliorated by prefrontal PCP (0.1mM) treatment whereas PPI was not affected by prefrontal cortex PCP administration per se at any of the doses tested. Taken together, inhibition of NMDA receptors in the PFC does not seem to be enough to impair PPI per se but NMDA receptor mediated signalling in the PFC may be a key factor for the PPI-disruptive effects of global NMDA receptor inhibition. This indicates that targeting PFC NMDA receptor signalling may have potential as a treatment target for schizophrenia although further studies are needed to understand pharmacology and pathophysiological role of PFC NMDA receptors.


Asunto(s)
Cognición/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Fenciclidina/farmacología , Corteza Prefrontal/fisiopatología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Esquizofrenia/fisiopatología , Filtrado Sensorial/efectos de los fármacos , Animales , Ácido Glutámico/metabolismo , Inyecciones Intraperitoneales , Masculino , Ratones , Corteza Prefrontal/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
13.
Behav Brain Res ; 213(2): 142-7, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20417666

RESUMEN

The use of translational approaches to validate animal models is needed for the development of treatments that can effectively alleviate cognitive impairments associated with schizophrenia, which are unsuccessfully treated by the current available therapies. Deficits in pre-attentive stages of sensory information processing seen in schizophrenia patients, can be assessed by highly homologues methods in both humans and rodents, evident by the prepulse inhibition (PPI) of the auditory startle response and the P50 (termed P1 here) suppression paradigms. Treatment with the NMDA receptor antagonist PCP on postnatal days 7, 9, and 11 reliably induce cognitive impairments resembling those presented by schizophrenia patients. Here we evaluate the potential of early postnatal PCP (20mg/kg) treatment in Lister Hooded rats to induce post-pubertal deficits in PPI and changes, such as reduced gating, in the P1 suppression paradigm in the EEG. The results indicate that early postnatal PCP treatment to rats leads to a reduction in PPI of the acoustic startle response. Furthermore, treated animals were assessed in the P1 suppression paradigm and produced significant changes in auditory-evoked potentials (AEP), specifically by an increased P1 amplitude and reduced P2 (P200 in humans) gating. However, the treatment neither disrupted normal P1 gating nor reduced N1 (N100 in humans) amplitude, representing two phenomena that are usually found to be disturbed in schizophrenia. In conclusion, the current findings confirm measures of early information processing to show high resemblance between rodents and humans, and indicate that early postnatal PCP-treated rats show deficits in pre-attentional processing, which are distinct from those observed in schizophrenia patients.


Asunto(s)
Modelos Animales de Enfermedad , Potenciales Evocados Auditivos/fisiología , Esquizofrenia/fisiopatología , Filtrado Sensorial/fisiología , Animales , Animales Recién Nacidos , Fenciclidina , Ratas , Ratas Endogámicas
14.
Synapse ; 63(12): 1083-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19642219

RESUMEN

Phencyclidine exerts psychotomimetic effects in humans and is used as a pharmacological animal model for schizophrenia. We, and others, have demonstrated that phencyclidine induces cognitive deficits in rats that are associated with schizophrenia. These cognitive deficits can be normalized by inhibition of nitric oxide synthase. The development of selective microelectrochemical nitric oxide sensors may provide direct evidence for the involvement of nitric oxide in these effects. The aim of the present study was to use LIVE (long term in vivo electrochemistry) to investigate the effect of phencyclidine, alone or in combination with the nitric oxide synthase inhibitor L-NAME, on nitric oxide levels in the medial prefrontal cortex of freely moving rats. Phencyclidine (2 mg kg(-1)) produced an increase in cortical nitric oxide levels and this increase was ameliorated by L-NAME (10 mg kg(-1)). Tentatively, the results from the present study provide a biochemical rationale for the involvement of nitric oxide in the phencyclidine model of schizophrenia.


Asunto(s)
Alucinógenos/farmacología , Óxido Nítrico/metabolismo , Fenciclidina/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Análisis de Varianza , Animales , Inhibidores Enzimáticos/farmacología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Ratas Wistar
15.
J Neurosci ; 29(7): 2238-51, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19228977

RESUMEN

A major challenge in neuroscience is to resolve the connection between gene functionality, neuronal circuits, and behavior. Most, if not all, neuronal circuits of the adult brain contain a glutamatergic component, the nature of which has been difficult to assess because of the vast cellular abundance of glutamate. In this study, we wanted to determine the role of a restricted subpopulation of glutamatergic neurons within the forebrain, the Vglut2-expressing neurons, in neuronal circuitry of higher brain function. Vglut2 expression was selectively deleted in the cortex, hippocampus, and amygdala of preadolescent mice, which resulted in increased locomotor activity, altered social dominance and risk assessment, decreased sensorimotor gating, and impaired long-term spatial memory. Presynaptic VGLUT2-positive terminals were lost in the cortex, striatum, nucleus accumbens, and hippocampus, and a downstream effect on dopamine binding site availability in the striatum was evident. A connection between the induced late-onset, chronic reduction of glutamatergic neurotransmission and dopamine signaling within the circuitry was further substantiated by a partial attenuation of the deficits in sensorimotor gating by the dopamine-stabilizing antipsychotic drug aripiprazole and an increased sensitivity to amphetamine. Somewhat surprisingly, given the restricted expression of Vglut2 in regions responsible for higher brain function, our analyses show that VGLUT2-mediated neurotransmission is required for certain aspects of cognitive, emotional, and social behavior. The present study provides support for the existence of a neurocircuitry that connects changes in VGLUT2-mediated neurotransmission to alterations in the dopaminergic system with schizophrenia-like behavioral deficits as a major outcome.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Dopamina/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Envejecimiento/metabolismo , Amígdala del Cerebelo/crecimiento & desarrollo , Animales , Antipsicóticos/farmacología , Conducta Animal/fisiología , Diferenciación Celular/genética , Corteza Cerebral/crecimiento & desarrollo , Cuerpo Estriado/crecimiento & desarrollo , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Ácido Glutámico/metabolismo , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Ratones , Ratones Noqueados , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Plasticidad Neuronal/genética , Núcleo Accumbens/crecimiento & desarrollo , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Filtrado Sensorial/genética , Transmisión Sináptica/genética
16.
Neuropsychopharmacology ; 34(7): 1673-84, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19145229

RESUMEN

Recent theories propose that both GABA and glutamate signaling are compromised in patients with schizophrenia. These deficits can be observed in several brain regions including the prefrontal cortex (PFC), an area extensively linked to the cognitive dysfunction in this disease and notably affected by NMDA receptor antagonists such as phencyclidine (PCP). We have previously demonstrated that inhibition of the nitric oxide (NO) pathways in the brain, particularly in the PFC, prevents a wide range of PCP-induced behavioral deficits including disruption of prepulse inhibition (PPI). This study investigated the role of GABA(B) receptor signaling and NO in the effects of PCP on PPI. Mice received systemic or prefrontal injections of the GABA(B) receptor agonist baclofen (2.5-5 mg/kg and 1 mM) before PCP treatment (5 mg/kg) and were thereafter tested for PPI. GABA/NO interactions were studied by combining baclofen and the NO synthase inhibitor L-NAME (20 mg/kg) in subthreshold doses. The role of GABA(B) receptors for NO production in vivo was assessed using NO-sensors implanted into the rat PFC. PCP-induced PPI deficits were attenuated in an additive manner by systemic baclofen treatment, whereas prefrontal microinjections of baclofen completely blocked the effects of PCP, without affecting PPI per se. The combination of baclofen and L-NAME was more effective in preventing the effects of PCP than any compound by itself. Additionally, baclofen decreased NO release in the PFC in a dose-related manner. This study proposes a role for GABA(B) receptor signaling in the effects of PCP, with altered NO levels as a downstream consequence. Thus, prefrontal NO signaling mirrors an altered level of cortical inhibition that may be of importance for information processing deficits in schizophrenia.


Asunto(s)
Trastornos del Conocimiento , Inhibición Neural/fisiología , Óxido Nítrico/metabolismo , Fenciclidina , Corteza Prefrontal/metabolismo , Receptores de GABA-B/metabolismo , Estimulación Acústica/métodos , Animales , Baclofeno/farmacología , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Electroquímica/métodos , Inhibidores Enzimáticos/farmacología , Agonistas del GABA/farmacología , Masculino , Ratones , Ratones Endogámicos , NG-Nitroarginina Metil Éster/farmacología , Inhibición Neural/efectos de los fármacos , Fenciclidina/farmacología , Corteza Prefrontal/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
17.
Behav Brain Res ; 200(1): 113-6, 2009 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-19166879

RESUMEN

Schizophrenia is a chronic disorder generally considered to encompass positive symptoms, negative symptoms and cognitive deficits. Increasing attention has been paid to the social cognitive deficits of the disorder as these dysfunctions are particularly handicapping, predictive of functional outcome and show poor treatment response. Phencyclidine (PCP) is a psychotomimetic drug used to model the different aspects of schizophrenia in experimental animal models. PCP-induced cognitive deficits and hyperlocomotion may be blocked by pretreatment with nitric oxide (NO) synthase inhibitors in rodents. The present study was carried out to evaluate the acute effects of PCP and NO synthase inhibition on social interaction in male Sprague-Dawley rats. The NO synthase inhibitor, L-NAME (10mg/kg) and PCP (2mg/kg) was injected subcutaneously to rats, which were then tested in pairs for social interactive behaviour. Twenty-four hours after the initial test a drug-free social interaction test was carried out to assess the rats' memory of the previous social interaction. The results showed that PCP reduced the time of social interaction on Day 1 compared to controls, and that pretreatment with the NO synthase inhibitor, L-NAME, attenuated this reduction towards control levels. Neither locomotor activity, nor frequency of social interactions were affected by the PCP treatment, suggesting that the PCP-induced effects observed were not due to drug-induced stereotypies. In combination with increasing clinical evidence for the involvement of NO in the pathophysiology of schizophrenia, the present results indicate that NO synthase inhibition may be a potentially new treatment strategy for alleviating social dysfunction in schizophrenia.


Asunto(s)
Conducta Animal/fisiología , Relaciones Interpersonales , Óxido Nítrico/fisiología , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Esquema de Medicación , Inhibidores Enzimáticos/farmacología , Alucinógenos/farmacología , Inyecciones Subcutáneas/métodos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Fenciclidina/farmacología , Ratas , Ratas Sprague-Dawley
18.
Eur J Pharmacol ; 590(1-3): 212-6, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18573247

RESUMEN

Agmatine, a decarboxylation product of arginine, is thought to be an important neuromodulator in the mammalian brain. It is proposed to exert neuroprotective, anxiolytic and antidepressant effects. The receptor-binding profile of agmatine is complex and includes interaction with alpha(2)-adrenergic and imidazoline I(1) receptors. Furthermore, agmatine is an NMDA-receptor antagonist and inhibits nitric oxide synthase. Prepulse inhibition (PPI) of the acoustic startle response is used as a measure of the pre-attentive information processing. PPI is lowered in schizophrenia and this impairment can be mimicked in experimental animals using the psychotomimetic drug phencyclidine (PCP). The aim of the present study was to investigate the effects of agmatine per se on the PPI response and the effects of agmatine pre-treatment on a PCP-induced disruption of PPI. Agmatine administration (10, 20 and 40 mg/kg) did not change the PPI response or the acoustic startle response. However, pre-treatment with agmatine 20 mg/kg, but not agmatine 40 mg/kg, significantly attenuated a PCP (5 mg/kg)-induced disruption of the PPI response. These results emphasize the potential role of agmatine as a neuromodulator and potential target for novel treatments for brain disorders.


Asunto(s)
Agmatina/farmacología , Procesos Mentales/efectos de los fármacos , Fenciclidina/farmacología , Reflejo de Sobresalto/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Óxido Nítrico Sintasa/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
19.
Pharmacol Biochem Behav ; 89(3): 352-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18321564

RESUMEN

Schizophrenia encompasses, amongst other symptoms, a heavy load of cognitive dysfunctionality. Using the psychotomimetic agent, phencyclidine (PCP), we have previously found that PCP-induced disruptions of cognitive function in translational rodent models of schizophrenia are dependent on nitric oxide (NO) production. In the present study, male Sprague-Dawley rats were subjected to a Morris water maze task designed to assess cognitive flexibility (i.e. the ability to cope with an increasingly demanding cognitive task) by means of a "constant reversal learning paradigm". Experiments were conducted to evaluate the effects of the NO synthase inhibitor, L-NAME (10 mg/kg), on PCP-induced (2 mg/kg) impairments. Control animals significantly improved their learning over the first 3 consecutive days, whereas PCP-treated animals failed to show any significant learning. Pretreatment with L-NAME normalized the PCP-induced disruption of learning to control levels. These findings suggest that PCP-induced disruptions of cognitive flexibility (i.e. ability to modify behaviour according to an increasingly demanding cognitive task) are dependent upon NO production. These observations, together with accumulated clinical findings, suggest that the NO system is a potential treatment target for cognitive dysfunctions in schizophrenia.


Asunto(s)
Cognición/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Fenciclidina/farmacología , Animales , Masculino , Óxido Nítrico/biosíntesis , Ratas , Ratas Sprague-Dawley , Conducta Estereotipada/efectos de los fármacos , Natación
20.
Neuropsychopharmacology ; 33(8): 1874-83, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17895915

RESUMEN

The prefrontal cortex (PFC) is believed to play an important role in the cognitive impairments observed in schizophrenia and has also been shown to be involved in the modulation of prepulse inhibition (PPI), a measure of preattentive information processing that is impaired in schizophrenic individuals. Phencyclidine (PCP), a noncompetitive inhibitor of the NMDA receptor, exerts psychotomimetic effects in humans, disrupts PPI, and causes hypofrontality in rodents and monkeys. We have previously demonstrated that interfering with the production of nitric oxide (NO) can prevent a wide range of PCP-induced behavioral deficits, including PPI disruption. In the present study, the role of NO signaling for the behavioral and biochemical effects of PCP was further investigated. Dialysate from the medial PFC of mice receiving systemic treatment with PCP and/or the NO synthase inhibitor, N(G)-nitro-L-arginine methyl ester (L-NAME, 40 mg/kg), was analyzed for cGMP content. Furthermore, a specific inhibitor of NO-sensitive soluble guanylyl cyclase (sGC), 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ, 0.01-1 mM), was administered into the medial PFC of mice in combination with systemic injections of PCP, followed by PPI and locomotor activity testing. PCP (5 mg/kg) caused an increase in prefrontal cGMP that could be attenuated by pretreatment with the NO synthase inhibitor, L-NAME. Moreover, bilateral microinjection of the sGC inhibitor, ODQ, into the medial PFC of mice attenuated the disruption of PPI, but not the hyperlocomotion, caused by PCP. The present study shows that NO/sGC/cGMP signaling pathway in the medial PFC is involved in specific behavioral effects of PCP that may have relevance for the disabling cognitive dysfunction found in patients with schizophrenia.


Asunto(s)
Conducta Animal/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Óxido Nítrico/fisiología , Fenciclidina/farmacología , Corteza Prefrontal/fisiología , Transducción de Señal/fisiología , Animales , GMP Cíclico/metabolismo , Método Doble Ciego , Electrodos Implantados , Inhibidores Enzimáticos/farmacología , Masculino , Ratones , Microdiálisis , Actividad Motora/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Quinoxalinas/farmacología , Radioinmunoensayo , Reflejo de Sobresalto/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA