Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
J Pharmacol Toxicol Methods ; 127: 107504, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38678804

RESUMO

We developed a novel, stress-free blood sampling method for minipigs, allowing continuous cortisol monitoring over 24 h. Baseline cortisol levels exhibited both ultradian and diurnal rhythms. During nighttime, smaller ultradian rhythms overlaid a lower baseline cortisol, which increased in sleeping pigs before lights were turned on. Additionally, we developed an analytical tool based on the R package "pracma" to quantify ultradian peak and circadian components of the cortisol profiles. To validate our model, we investigated the effects of Verucerfont, a CRH receptor antagonist, and Venlafaxine, a serotonin-norepinephrine reuptake inhibitor. Verucerfont reduced cortisol levels during the first 9 h without affecting diurnal rhythm. Cortisol peak parameters decreased, with a 31% reduction in overall area under the curve (AUC) and a 38% reduction in ultradian average AUC. Ultradian peaks decreased from 7 to 4.5, with 34% lower amplitude. Venlafaxine maintained plasma concentrations within the targeted human effective range. This method enables us to enhance our understanding of cortisol regulation and provide valuable insights for the impact of investigation drugs on the diurnal and ultradian rhythms of cortisol.


Assuntos
Ritmo Circadiano , Hidrocortisona , Porco Miniatura , Cloridrato de Venlafaxina , Animais , Suínos , Hidrocortisona/sangue , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/fisiologia , Cloridrato de Venlafaxina/farmacologia , Ritmo Ultradiano/efeitos dos fármacos , Ritmo Ultradiano/fisiologia , Coleta de Amostras Sanguíneas/métodos , Área Sob a Curva , Masculino , Feminino
2.
Eur J Pharmacol ; 953: 175802, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37295763

RESUMO

Antipsychotic drugs of different chemical/pharmacological families show preferential dopamine (DA) D2 receptor (D2-R) vs. D1 receptor (D1-R) affinity, with the exception of clozapine, the gold standard of schizophrenia treatment, which shows a comparable affinity for both DA receptors. Here, we examined the ability of Lu AF35700 (preferential D1-R>D2-R antagonist), to reverse the alterations in thalamo-cortical activity induced by phencyclidine (PCP), used as a pharmacological model of schizophrenia. Lu AF35700 reversed the PCP-induced alteration of neuronal discharge and low frequency oscillation (LFO, 0.15-4 Hz) in thalamo-cortical networks. Likewise, Lu AF35700 prevented the increased c-fos mRNA expression induced by PCP in thalamo-cortical regions of awake rats. We next examined the contribution of D1-R and D2-R to the antipsychotic reversal of PCP effects. The D2-R antagonist haloperidol reversed PCP effects on thalamic discharge rate and LFO. Remarkably, the combination of sub-effective doses of haloperidol and SCH-23390 (DA D1-R antagonist) fully reversed the PCP-induced fall in thalamo-cortical LFO. However, unlike with haloperidol, SCH-23390 elicited different degrees of potentiation of the effects of low clozapine and Lu AF35700 doses. Overall, the present data support a synergistic interaction between both DA receptors to reverse the PCP-induced alterations of oscillatory activity in thalamo-cortical networks, possibly due to their simultaneous blockade in direct and indirect pathways of basal ganglia. The mild potentiation induced by SCH-23390 in the case of clozapine and Lu AF35700 suggests that, at effective doses, these agents reverse PCP effects through the simultaneous blockade of both DA receptors.


Assuntos
Antipsicóticos , Clozapina , Ratos , Animais , Fenciclidina/farmacologia , Clozapina/farmacologia , Haloperidol/farmacologia , Dopamina , Antipsicóticos/farmacologia , Antagonistas de Dopamina/farmacologia , Receptores de Dopamina D1
3.
Schizophr Bull ; 49(4): 881-892, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-36454006

RESUMO

BACKGROUND: It remains unknown why ~30% of patients with psychotic disorders fail to respond to treatment. Previous genomic investigations of treatment-resistant psychosis have been inconclusive, but some evidence suggests a possible link between rare disease-associated copy number variants (CNVs) and worse clinical outcomes in schizophrenia. Here, we identified schizophrenia-associated CNVs in patients with treatment-resistant psychotic symptoms and then compared the prevalence of these CNVs to previously published schizophrenia cases not selected for treatment resistance. METHODS: CNVs were identified using chromosomal microarray (CMA) and whole exome sequencing (WES) in 509 patients with treatment-resistant psychosis (a lack of clinical response to ≥3 adequate antipsychotic medication trials over at least 5 years of psychiatric hospitalization). Prevalence of schizophrenia-associated CNVs in this sample was compared to that in a previously published large schizophrenia cohort study. RESULTS: Integrating CMA and WES data, we identified 47 cases (9.2%) with at least one CNV of known or possible neuropsychiatric risk. 4.7% (n = 24) carried a known neurodevelopmental risk CNV. The prevalence of well-replicated schizophrenia-associated CNVs was 4.1%, with duplications of the 16p11.2 and 15q11.2-q13.1 regions, and deletions of the 22q11.2 chromosomal region as the most frequent CNVs. Pairwise loci-based analysis identified duplications of 15q11.2-q13.1 to be independently associated with treatment resistance. CONCLUSIONS: These findings suggest that CNVs may uniquely impact clinical phenotypes beyond increasing risk for schizophrenia and may potentially serve as biological entry points for studying treatment resistance. Further investigation will be necessary to elucidate the spectrum of phenotypic characteristics observed in adult psychiatric patients with disease-associated CNVs.


Assuntos
Transtornos Psicóticos , Esquizofrenia , Humanos , Estudos de Coortes , Variações do Número de Cópias de DNA/genética , Prevalência , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/epidemiologia , Transtornos Psicóticos/genética , Esquizofrenia/tratamento farmacológico , Esquizofrenia/epidemiologia , Esquizofrenia/genética , Predisposição Genética para Doença
4.
Bioorg Med Chem Lett ; 72: 128879, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35809818

RESUMO

The N-demethylation of zicronapine (7) and three of its deuterated analogs 8 - 10 has been studied in human in vitro metabolism systems. While the N-deuterio-methyl analog 8 did not behave differently from the parent in human liver microsomes, a significantly reduced rate of N-demethylation was observed as a consequence of benzene ring deuteration (compound 7vs.9). Additional deuteration of the N-methyl group, which as mentioned had shown no effect in isolation, further decreased the rate of the N-demethylation reaction (compound 10vs.9). This paper presents and discusses this unprecedented 'distal kinetic isotope effect' that was observed when incubating the test compounds with human liver microsomes or recombinant human CYP450 liver enzymes.


Assuntos
Sistema Enzimático do Citocromo P-450 , Microssomos Hepáticos , Sistema Enzimático do Citocromo P-450/metabolismo , Desmetilação , Deutério/metabolismo , Humanos , Cinética , Microssomos Hepáticos/metabolismo
5.
Neuroimage ; 243: 118520, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34455061

RESUMO

Copy number variations (CNV) involving multiple genes are ideal models to study polygenic neuropsychiatric disorders. Since 22q11.2 deletion is regarded as the most important single genetic risk factor for developing schizophrenia, characterizing the effects of this CNV on neural networks offers a unique avenue towards delineating polygenic interactions conferring risk for the disorder. We used a Df(h22q11)/+ mouse model of human 22q11.2 deletion to dissect gene expression patterns that would spatially overlap with differential resting-state functional connectivity (FC) patterns in this model (N = 12 Df(h22q11)/+ mice, N = 10 littermate controls). To confirm the translational relevance of our findings, we analyzed tissue samples from schizophrenia patients and healthy controls using machine learning to explore whether identified genes were co-expressed in humans. Additionally, we employed the STRING protein-protein interaction database to identify potential interactions between genes spatially associated with hypo- or hyper-FC. We found significant associations between differential resting-state connectivity and spatial gene expression patterns for both hypo- and hyper-FC. Two genes, Comt and Trmt2a, were consistently over-expressed across all networks. An analysis of human datasets pointed to a disrupted co-expression of these two genes in the brain in schizophrenia patients, but not in healthy controls. Our findings suggest that COMT and TRMT2A form a core genetic component implicated in differential resting-state connectivity patterns in the 22q11.2 deletion. A disruption of their co-expression in schizophrenia patients points out a prospective cause for the aberrance of brain networks communication in 22q11.2 deletion syndrome on a molecular level.


Assuntos
Catecol O-Metiltransferase/genética , Síndrome de DiGeorge/genética , Expressão Gênica , tRNA Metiltransferases/genética , Animais , Deleção Cromossômica , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Esquizofrenia/genética
6.
Transl Psychiatry ; 11(1): 74, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33495438

RESUMO

Attention deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neurodevelopmental disorder worldwide. Affected individuals present with hyperactivity, inattention, and cognitive deficits and display a characteristic paradoxical response to drugs affecting the dopaminergic system. However, the underlying pathophysiology of ADHD and how this relates to dopaminergic transmission remains to be fully understood. Sorcs2-/- mice uniquely recapitulate symptoms reminiscent of ADHD in humans. Here, we show that lack of SorCS2 in mice results in lower sucrose intake, indicating general reward deficits. Using in-vivo recordings, we further find that dopaminergic transmission in the ventral tegmental area (VTA) is shifted towards a more regular firing pattern with marked reductions in the relative occurrence of irregular firing in Sorcs2-/- mice. This was paralleled by abnormal acute behavioral responses to dopamine receptor agonists, suggesting fundamental differences in dopaminergic circuits and indicating a perturbation in the balance between the activities of the postsynaptic dopamine receptor DRD1 and the presynaptic inhibitory autoreceptor DRD2. Interestingly, the hyperactivity and drug response of Sorcs2-/- mice were markedly affected by novelty. Taken together, our findings show how loss of a candidate ADHD-risk gene has marked effects on dopaminergic circuit function and the behavioral response to the environment.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Animais , Transtorno do Deficit de Atenção com Hiperatividade/genética , Dopamina , Camundongos , Proteínas do Tecido Nervoso , Receptores de Superfície Celular , Receptores Dopaminérgicos , Recompensa , Área Tegmentar Ventral
7.
Mol Psychiatry ; 26(5): 1520-1534, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-31705054

RESUMO

Genetic risk for psychiatric illness is complex, so identification of shared molecular pathways where distinct forms of genetic risk might coincide is of substantial interest. A growing body of genetic and genomic studies suggest that such shared molecular pathways exist across disorders with different clinical presentations, such as schizophrenia and autism spectrum disorder (ASD). But how this relates to specific genetic risk factors is unknown. Further, whether some of the molecular changes identified in brain relate to potentially confounding antemortem or postmortem factors are difficult to prove. We analyzed the transcriptome from the cortex and hippocampus of three mouse lines modeling human copy number variants (CNVs) associated with schizophrenia and ASD: Df(h15q13)/+, Df(h22q11)/+, and Df(h1q21)/+ which carry the 15q13.3 deletion, 22q11.2 deletion, and 1q21.1 deletion, respectively. Although we found very little overlap of differential expression at the level of individual genes, gene network analysis identified two cortical and two hippocampal modules of co-expressed genes that were dysregulated across all three mouse models. One cortical module was associated with neuronal energetics and firing rate, and overlapped with changes identified in postmortem human brain from SCZ and ASD patients. These data highlight aspects of convergent gene expression in mouse models harboring major risk alleles, and strengthen the connection between changes in neuronal energetics and neuropsychiatric disorders in humans.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Esquizofrenia , Animais , Transtorno do Espectro Autista/genética , Transtorno Autístico/genética , Deleção Cromossômica , Humanos , Camundongos , Esquizofrenia/genética , Transcriptoma/genética
8.
Transl Psychiatry ; 10(1): 239, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32681022

RESUMO

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.


Assuntos
Histona Acetiltransferases , Esquizofrenia , Animais , Depressão/genética , Epigênese Genética , Feminino , Expressão Gênica , Camundongos , Esquizofrenia/genética
9.
Transl Psychiatry ; 10(1): 93, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170065

RESUMO

22q11.2, 15q13.3, and 1q21.1 microdeletions attract considerable interest by conferring high risk for a range of neuropsychiatric disorders, including schizophrenia and autism. A fundamental open question is whether divergent or convergent neural mechanisms mediate this genetic pleiotropic association with the same behavioral phenotypes. We use a combination of rodent microdeletion models with high-field neuroimaging to perform a comparative whole-brain characterization of functional and structural mechanisms linked to high-risk states. Resting-state functional and structural magnetic resonance imaging data were acquired on mice carrying heterozygous microdeletions in 22q11.2 (N = 12), 15q13.3 (N = 11), and 1q21.1 (N = 11) loci. We performed network-based statistic, graph, and morphometric analyses. The three microdeletions did not share significant systems-level features. Instead, morphometric analyses revealed microcephaly in 1q21.1 and macrocephaly in 15q13.3 deletions, whereas cerebellar volume was specifically reduced in 22q11.2 deletion. In function, 22q11.2 deletion mice showed widespread cortical hypoconnectivity, accompanied by opposing hyperconnectivity in dopaminergic pathways, which was confirmed by graph analysis. 1q21.1 exhibited distinct changes in posterior midbrain morphology and function, especially in periaqueductal gray, whereas 15q13.3 demonstrated alterations in auditory/striatal system. The combination of cortical hypoconnectivity and dopaminergic hyperconnectivity and reduced cerebellum in 22q11.2 deletion mirrors key neurodevelopmental features of schizophrenia, whereas changes in midbrain and auditory/striatal morphology and topology in 1q21.1 and 15q13.3 rather indicate focal processes possibly linked to the emergence of abnormal salience perception and hallucinations. In addition to insights into pathophysiological processes in these microdeletions, our results establish the general point that microdeletions might increase risk for overlapping neuropsychiatric phenotypes through separable neural mechanisms.


Assuntos
Variações do Número de Cópias de DNA , Esquizofrenia , Animais , Encéfalo/diagnóstico por imagem , Deleção Cromossômica , Camundongos , Fenótipo , Esquizofrenia/diagnóstico por imagem , Esquizofrenia/genética
10.
Transl Psychiatry ; 10(1): 41, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-32066701

RESUMO

Genetic microdeletion at the 22q11 locus is associated with very high risk for schizophrenia. The 22q11.2 microdeletion (Df(h22q11)/+) mouse model shows cognitive deficits observed in this disorder, some of which can be linked to dysfunction of the prefrontal cortex (PFC). We used behavioral (n = 10 per genotype), electrophysiological (n = 7 per genotype per group), and neuroanatomical (n = 5 per genotype) techniques to investigate schizophrenia-related pathology of Df(h22q11)/+ mice, which showed a significant decrease in the total number of parvalbumin positive interneurons in the medial PFC. The Df(h22q11)/+ mice when tested on PFC-dependent behavioral tasks, including gambling tasks, perform significantly worse than control animals while exhibiting normal behavior on hippocampus-dependent tasks. They also show a significant decrease in hippocampus-medial Prefrontal cortex (H-PFC) synaptic plasticity (long-term potentiation, LTP). Acute platform stress almost abolished H-PFC LTP in both wild-type and Df(h22q11)/+ mice. H-PFC LTP was restored to prestress levels by clozapine (3 mg/kg i.p.) in stressed Df(h22q11)/+ mice, but the restoration of stress-induced LTP, while significant, was similar between wild-type and Df(h22q11)/+ mice. A medial PFC dysfunction may underlie the negative and cognitive symptoms in human 22q11 deletion carriers, and these results are relevant to the current debate on the utility of clozapine in such subjects.


Assuntos
Cognição , Córtex Pré-Frontal , Animais , Modelos Animais de Doenças , Hipocampo , Camundongos , Camundongos Endogâmicos C57BL
11.
PLoS One ; 14(6): e0217765, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31206532

RESUMO

BACKGROUND: Of the 108 Schizophrenia (SZ) risk-loci discovered through genome-wide association studies (GWAS), 96 are not altering the sequence of any protein. Evidence linking non-coding risk-SNPs and genes may be established using expression quantitative trait loci (eQTL). However, other approaches such allelic expression quantitative trait loci (aeQTL) also may be of use. METHODS: We applied both the eQTL and aeQTL analysis to a biobank of deeply sequenced RNA from 680 dorso-lateral pre-frontal cortex (DLPFC) samples. For each of 340 genes proximal to the SZ risk-SNPs, we asked how much SNP-genotype affected total expression (eQTL), as well as how much the expression ratio between the two alleles differed from 1:1 as a consequence of the risk-SNP genotype (aeQTL). RESULTS: We analyzed overlap with comparable eQTL-findings: 16 of the 30 risk-SNPs known to have gene-level eQTL also had gene-level aeQTL effects. 6 of 21 risk-SNPs with known splice-eQTL had exon-aeQTL effects. 12 novel potential risk genes were identified with the aeQTL approach, while 55 tested SNP-pairs were found as eQTL but not aeQTL. Of the tested 108 loci we could find at least one gene to be associated with 21 of the risk-SNPs using gene-level aeQTL, and with an additional 18 risk-SNPs using exon-level aeQTL. CONCLUSION: Our results suggest that the aeQTL strategy complements the eQTL approach to susceptibility gene identification.


Assuntos
Desequilíbrio Alélico/genética , Encéfalo/patologia , Estudo de Associação Genômica Ampla/métodos , Locos de Características Quantitativas/genética , RNA-Seq/métodos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença/genética , Genótipo , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Esquizofrenia/genética , Sequenciamento do Exoma/métodos , Adulto Jovem
12.
Neuroimage Clin ; 22: 101721, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30785050

RESUMO

One of the most common copy number variants, the 22q11.2 microdeletion, confers an increased risk for schizophrenia. Since schizophrenia has been associated with an aberrant neural response to repeated stimuli through both reduced adaptation and prediction, we here hypothesized that this may also be the case in nonpsychotic individuals with a 22q11.2 deletion. We recorded high-density EEG from 19 individuals with 22q11.2 deletion syndrome (12-25 years), as well as 27 healthy volunteers with comparable age and sex distribution, while they listened to a sequence of sounds arranged in a roving oddball paradigm. Using posterior probability maps and dynamic causal modelling we tested three different models accounting for repetition dependent changes in cortical responses as well as in effective connectivity; namely an adaptation model, a prediction model, and a model including both adaptation and prediction. Repetition-dependent changes were parametrically modulated by a combination of adaptation and prediction and were apparent in both cortical responses and in the underlying effective connectivity. This effect was reduced in individuals with a 22q11.2 deletion and was negatively correlated with negative symptom severity. Follow-up analysis showed that the reduced effect of the combined adaptation and prediction model seen in individuals with 22q11.2 deletion was driven by reduced adaptation rather than prediction failure. Our findings suggest that adaptation is reduced in individuals with a 22q11.2 deletion, which can be interpreted in light of the framework of predictive coding as a failure to suppress prediction errors.


Assuntos
Síndrome da Deleção 22q11/fisiopatologia , Adaptação Fisiológica/fisiologia , Percepção Auditiva/fisiologia , Encéfalo/fisiopatologia , Estimulação Acústica , Adolescente , Adulto , Teorema de Bayes , Criança , Eletroencefalografia , Feminino , Humanos , Masculino , Adulto Jovem
13.
Neuropsychopharmacology ; 44(4): 703-710, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30188511

RESUMO

15q13.3 microdeletion is one of several gene copy number variants (CNVs) conferring increased risk of psychiatric and neurological disorders. This microdeletion gives rise to a variable spectrum of pathological phenotypes, ranging from asymptomatic to severe clinical outcomes. The reasons for these varying phenotypic outcomes remain unknown. Using a mouse model of hemizygous deletion of the orthologous region of 15q13.3, the present study examined whether exposure to stressful life events might interact with hemizygous 15q13.3 microdeletion in the development of behavioral dysfunctions. We show that hemizygous 15q13.3 microdeletion alone induces only limited effects on adult behaviors, but when combined with psychological stress in pubescence (postnatal days 30-40), it impairs sensorimotor gating and increases the sensitivity to the psychostimulant drug, amphetamine, at adult age. Stress exposure in adolescence (postnatal days 50-60) did not induce similar interactions with 15q13.3 microdeletion, but led to impaired emotional learning and memory and social behavior regardless of the genetic background. The present study provides the first evidence for interactive effects between hemizygous 15q13.3 microdeletion and exposure to stressful life events, and at the same time, it emphasizes an important influence of the precise timing of postnatal stress exposure in these interactions. Our findings suggest that hemizygous 15q13.3 microdeletion can act as a "disease primer" that increases the carrier's vulnerability to the detrimental effects of peripubertal stress exposure on adult behaviors.


Assuntos
Anedonia/fisiologia , Ansiedade/fisiopatologia , Transtornos Cromossômicos/fisiopatologia , Emoções/fisiologia , Deficiência Intelectual/fisiopatologia , Aprendizagem/fisiologia , Convulsões/fisiopatologia , Filtro Sensorial/fisiologia , Comportamento Social , Estresse Psicológico/fisiopatologia , Fatores Etários , Anfetamina/farmacologia , Animais , Comportamento Animal/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Deleção Cromossômica , Transtornos Cromossômicos/complicações , Cromossomos Humanos Par 15 , Modelos Animais de Doenças , Hemizigoto , Deficiência Intelectual/complicações , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/complicações , Filtro Sensorial/efeitos dos fármacos , Maturidade Sexual/fisiologia
14.
Biol Psychiatry ; 85(1): 13-24, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30144930

RESUMO

The diagnosis of schizophrenia rests on clinical criteria that cannot be assessed in animal models. Together with absence of a clear underlying pathology and understanding of what causes schizophrenia, this has hindered development of informative animal models. However, recent large-scale genomic studies have identified copy number variants (CNVs) that confer high risk of schizophrenia and have opened a new avenue for generation of relevant animal models. Eight recurrent CNVs have reproducibly been shown to increase the risk of schizophrenia by severalfold: 22q11.2(del), 15q13.3(del), 1q21(del), 1q21(dup), NRXN1(del), 3q29(del), 7q11.23(dup), and 16p11.2(dup). Five of these CNVs have been modeled in animals, mainly mice, but also rats, flies, and zebrafish, and have been shown to recapitulate behavioral and electrophysiological aspects of schizophrenia. Here, we provide an overview of the schizophrenia-related phenotypes found in animal models of schizophrenia high-risk CNVs. We also discuss strengths and limitations of the CNV models, and how they can advance our biological understanding of mechanisms that can lead to schizophrenia and can be used to develop new and better treatments for schizophrenia.


Assuntos
Transtornos Psicóticos , Esquizofrenia , Animais , Proteínas de Ligação ao Cálcio , Variações do Número de Cópias de DNA , Dopamina , Camundongos , Modelos Animais , Moléculas de Adesão de Célula Nervosa , Tomografia por Emissão de Pósitrons , Ratos
15.
Transl Psychiatry ; 8(1): 247, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30429456

RESUMO

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.


Assuntos
Atenção/fisiologia , Comportamento Animal/fisiologia , Estimulantes do Sistema Nervoso Central/farmacologia , Disfunção Cognitiva/fisiopatologia , Síndrome de DiGeorge/fisiopatologia , Sincronização de Fases em Eletroencefalografia/fisiologia , Função Executiva/fisiologia , Hipocampo/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Desempenho Psicomotor/fisiologia , Anfetamina/farmacologia , Animais , Atenção/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Função Executiva/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Modafinila/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos
16.
Schizophr Res ; 197: 328-336, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29395612

RESUMO

22q11.2 deletion syndrome (22q11.2DS) is one of the most common copy number variants and confers a markedly increased risk for schizophrenia. As such, 22q11.2DS is a homogeneous genetic liability model which enables studies to delineate functional abnormalities that may precede disease onset. Mismatch negativity (MMN), a brain marker of change detection, is reduced in people with schizophrenia compared to healthy controls. Using dynamic causal modelling (DCM), previous studies showed that top-down effective connectivity linking the frontal and temporal cortex is reduced in schizophrenia relative to healthy controls in MMN tasks. In the search for early risk-markers for schizophrenia we investigated the neural basis of change detection in a group with 22q11.2DS. We recorded high-density EEG from 19 young non-psychotic 22q11.2 deletion carriers, as well as from 27 healthy non-carriers with comparable age distribution and sex ratio, while they listened to a sequence of sounds arranged in a roving oddball paradigm. Despite finding no significant reduction in the MMN responses, whole-scalp spatiotemporal analysis of responses to the tones revealed a greater fronto-temporal N1 component in the 22q11.2 deletion carriers. DCM showed reduced intrinsic connection within right primary auditory cortex as well as in the top-down, connection from the right inferior frontal gyrus to right superior temporal gyrus for 22q11.2 deletion carriers although not surviving correction for multiple comparison. We discuss these findings in terms of reduced adaptation and a general increased sensitivity to tones in 22q11.2DS.


Assuntos
Percepção Auditiva/fisiologia , Síndrome de DiGeorge/fisiopatologia , Potenciais Evocados Auditivos/fisiologia , Córtex Pré-Frontal/fisiopatologia , Lobo Temporal/fisiopatologia , Adolescente , Adulto , Córtex Auditivo/fisiopatologia , Criança , Eletroencefalografia , Feminino , Heterozigoto , Humanos , Masculino , Modelos Teóricos , Análise Espaço-Temporal , Adulto Jovem
17.
Schizophr Bull ; 44(2): 388-397, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28521049

RESUMO

Background: The 22q11.2 deletion syndrome confers a markedly increased risk for schizophrenia. 22q11.2 deletion carriers without manifest psychotic disorder offer the possibility to identify functional abnormalities that precede clinical onset. Since schizophrenia is associated with a reduced cortical gamma response to auditory stimulation at 40 Hz, we hypothesized that the 40 Hz auditory steady-state response (ASSR) may be attenuated in nonpsychotic individuals with a 22q11.2 deletion. Methods: Eighteen young nonpsychotic 22q11.2 deletion carriers and a control group of 27 noncarriers with comparable age range (12-25 years) and sex ratio underwent 128-channel EEG. We recorded the cortical ASSR to a 40 Hz train of clicks, given either at a regular inter-stimulus interval of 25 ms or at irregular intervals jittered between 11 and 37 ms. Results: Healthy noncarriers expressed a stable ASSR to regular but not in the irregular 40 Hz click stimulation. Both gamma power and inter-trial phase coherence of the ASSR were markedly reduced in the 22q11.2 deletion group. The ability to phase lock cortical gamma activity to regular auditory 40 Hz stimulation correlated with the individual expression of negative symptoms in deletion carriers (ρ = -0.487, P = .041). Conclusions: Nonpsychotic 22q11.2 deletion carriers lack efficient phase locking of evoked gamma activity to regular 40 Hz auditory stimulation. This abnormality indicates a dysfunction of fast intracortical oscillatory processing in the gamma-band. Since ASSR was attenuated in nonpsychotic deletion carriers, ASSR deficiency may constitute a premorbid risk marker of schizophrenia.


Assuntos
Córtex Auditivo/fisiopatologia , Percepção Auditiva/fisiologia , Síndrome de DiGeorge/fisiopatologia , Eletroencefalografia/métodos , Potenciais Evocados Auditivos/fisiologia , Ritmo Gama/fisiologia , Adolescente , Criança , Feminino , Humanos , Masculino , Adulto Jovem
18.
Transl Psychiatry ; 7(11): 1261, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29187755

RESUMO

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.


Assuntos
Anormalidades Múltiplas , Comportamento Animal , Deleção Cromossômica , Agonistas de Dopamina/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Neurônios Dopaminérgicos/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Megalencefalia , Núcleo Accumbens/metabolismo , Inibição Pré-Pulso , Receptores Dopaminérgicos/metabolismo , Esquizofrenia , Área Tegmentar Ventral/metabolismo , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Anormalidades Múltiplas/fisiopatologia , Anfetamina/farmacologia , Animais , Apomorfina/farmacologia , Comportamento Animal/efeitos dos fármacos , Benzazepinas/farmacologia , Cromossomos Humanos Par 1/metabolismo , Modelos Animais de Doenças , Agonistas de Dopamina/administração & dosagem , Inibidores da Captação de Dopamina/administração & dosagem , Neurônios Dopaminérgicos/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Megalencefalia/metabolismo , Megalencefalia/patologia , Megalencefalia/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Accumbens/efeitos dos fármacos , Fenciclidina/farmacologia , Fenótipo , Inibição Pré-Pulso/efeitos dos fármacos , Quimpirol/farmacologia , Receptores Dopaminérgicos/efeitos dos fármacos , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Esquizofrenia/fisiopatologia , Área Tegmentar Ventral/efeitos dos fármacos
19.
CNS Neurosci Ther ; 23(6): 518-525, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28417559

RESUMO

AIM: Asenapine is a new atypical antipsychotic prescribed for the treatment of psychosis/bipolar disorders that presents higher affinity for serotonergic than dopaminergic receptors. The objective of this study was to investigate its antidepressant-like and antimanic-like properties on relevant animal models of depression and mania and to assess the acute and chronic effect of Asenapine on dorsal raphe nucleus (DRN) 5-HT cell firing activity. METHODS: We assessed the effects of Asenapine using in vivo electrophysiological and behavioral assays in rats. RESULTS: Behavioral experiments showed that Asenapine had no significant effect on immobility time in the forced swim test (FST) in control rats. In the ACTH-treated rats, a model of antidepressant-resistance, Asenapine failed to alter immobility time in the FST. In contrast in the sleep deprivation (SD) model of mania, acute administration of Asenapine significantly decreased the hyperlocomotion of SD rats. In the DRN, acute administration of Asenapine reduced the suppressant effect of the selective 5-HT7 receptor agonist LP-44 and of the prototypical 5-HT1A receptor agonist 8-OH-DPAT on 5-HT neuronal firing activity. In addition, chronic treatment with Asenapine enhanced DRN 5-HT neuronal firing and this effect was associated with an alteration of the 5-HT7 receptor responsiveness. CONCLUSION: These results confirm that Asenapine displays robust antimanic property and effective in vivo antagonistic activity at 5-HT1A/7 receptors.


Assuntos
Afeto/efeitos dos fármacos , Antipsicóticos/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Receptor 5-HT1A de Serotonina/metabolismo , Receptores de Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Hormônio Adrenocorticotrópico/farmacologia , Afeto/fisiologia , Animais , Dexmedetomidina/farmacologia , Dibenzocicloeptenos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Masculino , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Núcleos Septais/citologia , Serotoninérgicos/farmacologia , Privação do Sono/tratamento farmacológico , Privação do Sono/metabolismo , Natação/psicologia
20.
Neurobiol Learn Mem ; 141: 44-52, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28341151

RESUMO

Schizophrenia is a debilitating brain disorder characterized by disturbances of emotion, perception and cognition. Cognitive impairments predict functional outcome in schizophrenia and are detectable even in the prodromal stage of the disorder. However, our understanding of the underlying neurobiology is limited and procognitive treatments remain elusive. We recently demonstrated that mice heterozygous for an inactivated allele of the schizophrenia-associated Brd1 gene (Brd1+/- mice) display behaviors reminiscent of schizophrenia, including impaired social cognition and long-term memory. Here, we further characterize performance of these mice by following the preclinical guidelines recommended by the 'Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS)' and 'Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia (CNTRICS)' initiatives to maximize translational value. Brd1+/- mice exhibit relational encoding deficits, compromised working and long term memory, as well as impaired executive cognitive functioning with cognitive behaviors relying on medial prefrontal cortex being particularly affected. Akin to patients with schizophrenia, the cognitive deficits displayed by Brd1+/- mice are not global, but selective. Our results underline the value of Brd1+/- mice as a promising tool for studying the neurobiology of cognitive deficits in schizophrenia.


Assuntos
Transtornos Cognitivos/genética , Cognição/fisiologia , Função Executiva/fisiologia , Histona Acetiltransferases/genética , Esquizofrenia/genética , Alelos , Animais , Comportamento Animal/fisiologia , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Heterozigoto , Masculino , Memória de Longo Prazo/fisiologia , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Knockout , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...