Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Cell ; 152(1-2): 262-75, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23332760

RESUMO

22q11.2 microdeletions result in specific cognitive deficits and schizophrenia. Analysis of Df(16)A(+/-) mice, which model this microdeletion, revealed abnormalities in the formation of neuronal dendrites and spines, as well as altered brain microRNAs. Here, we show a drastic reduction of miR-185, which resides within the 22q11.2 locus, to levels more than expected by a hemizygous deletion, and we demonstrate that this reduction alters dendritic and spine development. miR-185 represses, through an evolutionarily conserved target site, a previously unknown inhibitor of these processes that resides in the Golgi apparatus and shows higher prenatal brain expression. Sustained derepression of this inhibitor after birth represents the most robust transcriptional disturbance in the brains of Df(16)A(+/-) mice and results in structural alterations in the hippocampus. Reduction of miR-185 also has milder age- and region-specific effects on the expression of some Golgi-related genes. Our findings illuminate the contribution of microRNAs in psychiatric disorders and cognitive dysfunction.


Assuntos
Encéfalo/metabolismo , Embrião de Mamíferos/metabolismo , MicroRNAs/metabolismo , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Modelos Animais de Doenças , Complexo de Golgi/metabolismo , Hipocampo/metabolismo , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , MicroRNAs/genética , Dados de Sequência Molecular , Plasticidade Neuronal , Neurônios/metabolismo , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Proteínas de Ligação a RNA
2.
Nature ; 591(7851): 615-619, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33627872

RESUMO

The ability to rapidly adapt to novel situations is essential for survival, and this flexibility is impaired in many neuropsychiatric disorders1. Thus, understanding whether and how novelty prepares, or primes, brain circuitry to facilitate cognitive flexibility has important translational relevance. Exposure to novelty recruits the hippocampus and medial prefrontal cortex (mPFC)2 and may prime hippocampal-prefrontal circuitry for subsequent learning-associated plasticity. Here we show that novelty resets the neural circuits that link the ventral hippocampus (vHPC) and the mPFC, facilitating the ability to overcome an established strategy. Exposing mice to novelty disrupted a previously encoded strategy by reorganizing vHPC activity to local theta (4-12 Hz) oscillations and weakening existing vHPC-mPFC connectivity. As mice subsequently adapted to a new task, vHPC neurons developed new task-associated activity, vHPC-mPFC connectivity was strengthened, and mPFC neurons updated to encode the new rules. Without novelty, however, mice adhered to their established strategy. Blocking dopamine D1 receptors (D1Rs) or inhibiting novelty-tagged cells that express D1Rs in the vHPC prevented these behavioural and physiological effects of novelty. Furthermore, activation of D1Rs mimicked the effects of novelty. These results suggest that novelty promotes adaptive learning by D1R-mediated resetting of vHPC-mPFC circuitry, thereby enabling subsequent learning-associated circuit plasticity.


Assuntos
Hipocampo/fisiologia , Aprendizagem em Labirinto/fisiologia , Vias Neurais/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Córtex Pré-Frontal/fisiologia , Animais , Feminino , Hipocampo/citologia , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/citologia
3.
Nature ; 522(7556): 309-14, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-26053122

RESUMO

Spatial working memory, the caching of behaviourally relevant spatial cues on a timescale of seconds, is a fundamental constituent of cognition. Although the prefrontal cortex and hippocampus are known to contribute jointly to successful spatial working memory, the anatomical pathway and temporal window for the interaction of these structures critical to spatial working memory has not yet been established. Here we find that direct hippocampal-prefrontal afferents are critical for encoding, but not for maintenance or retrieval, of spatial cues in mice. These cues are represented by the activity of individual prefrontal units in a manner that is dependent on hippocampal input only during the cue-encoding phase of a spatial working memory task. Successful encoding of these cues appears to be mediated by gamma-frequency synchrony between the two structures. These findings indicate a critical role for the direct hippocampal-prefrontal afferent pathway in the continuous updating of task-related spatial information during spatial working memory.


Assuntos
Hipocampo/fisiologia , Memória de Curto Prazo/fisiologia , Córtex Pré-Frontal/fisiologia , Percepção Espacial/fisiologia , Memória Espacial/fisiologia , Potenciais de Ação , Vias Aferentes/fisiologia , Animais , Sinais (Psicologia) , Hipocampo/citologia , Masculino , Camundongos , Modelos Neurológicos , Optogenética , Córtex Pré-Frontal/citologia
4.
J Neurosci ; 39(18): 3561-3581, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30833507

RESUMO

Neurodevelopmental disorders offer insight into synaptic mechanisms. To unbiasedly uncover these mechanisms, we studied the 22q11.2 syndrome, a recurrent copy number variant, which is the highest schizophrenia genetic risk factor. We quantified the proteomes of 22q11.2 mutant human fibroblasts from both sexes and mouse brains carrying a 22q11.2-like defect, Df(16)A+/- Molecular ontologies defined mitochondrial compartments and pathways as some of top ranked categories. In particular, we identified perturbations in the SLC25A1-SLC25A4 mitochondrial transporter interactome as associated with the 22q11.2 genetic defect. Expression of SLC25A1-SLC25A4 interactome components was affected in neuronal cells from schizophrenia patients. Furthermore, hemideficiency of the Drosophila SLC25A1 or SLC25A4 orthologues, dSLC25A1-sea and dSLC25A4-sesB, affected synapse morphology, neurotransmission, plasticity, and sleep patterns. Our findings indicate that synapses are sensitive to partial loss of function of mitochondrial solute transporters. We propose that mitoproteomes regulate synapse development and function in normal and pathological conditions in a cell-specific manner.SIGNIFICANCE STATEMENT We address the central question of how to comprehensively define molecular mechanisms of the most prevalent and penetrant microdeletion associated with neurodevelopmental disorders, the 22q11.2 microdeletion syndrome. This complex mutation reduces gene dosage of ∼63 genes in humans. We describe a disruption of the mitoproteome in 22q11.2 patients and brains of a 22q11.2 mouse model. In particular, we identify a network of inner mitochondrial membrane transporters as a hub required for synapse function. Our findings suggest that mitochondrial composition and function modulate the risk of neurodevelopmental disorders, such as schizophrenia.


Assuntos
Síndrome da Deleção 22q11/metabolismo , Encéfalo/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Translocador 1 do Nucleotídeo Adenina/metabolismo , Animais , Comportamento Animal , Linhagem Celular , Deleção Cromossômica , Cromossomos Humanos Par 22/metabolismo , Drosophila , Feminino , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas Mitocondriais/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Proteoma , Esquizofrenia/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(30): E6127-E6136, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28696314

RESUMO

Identification of protective loss-of-function (LoF) mutations holds great promise for devising novel therapeutic interventions, although it faces challenges due to the scarcity of protective LoF alleles in the human genome. Exploiting the detailed mechanistic characterization of animal models of validated disease mutations offers an alternative. Here, we provide insights into protective-variant biology based on our characterization of a model of the 22q11.2 deletion, a strong genetic risk factor for schizophrenia (SCZ). Postnatal brain up-regulation of Mirta22/Emc10, an inhibitor of neuronal maturation, represents the major transcriptional effect of the 22q11.2-associated microRNA dysregulation. Here, we demonstrate that mice in which the Df(16)A deficiency is combined with a LoF Mirta22 allele show rescue of key SCZ-related deficits, namely prepulse inhibition decrease, working memory impairment, and social memory deficits, as well as synaptic and structural plasticity abnormalities in the prefrontal cortex. Additional analysis of homozygous Mirta22 knockout mice, in which no alteration is observed in the above-mentioned SCZ-related phenotypes, highlights the deleterious effects of Mirta22 up-regulation. Our results support a causal link between dysregulation of a miRNA target and SCZ-related deficits and provide key insights into beneficial LoF mutations and potential new treatments.


Assuntos
Mutação com Perda de Função , Proteínas de Membrana/genética , Esquizofrenia/genética , Animais , Feminino , Predisposição Genética para Doença , Masculino , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/genética , Fenótipo , Esquizofrenia/fisiopatologia
6.
Cereb Cortex ; 28(6): 2175-2191, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28525574

RESUMO

Altered prefrontal cortex function is implicated in schizophrenia (SCZ) pathophysiology and could arise from imbalance between excitation and inhibition (E/I) in local circuits. It remains unclear whether and how such imbalances relate to genetic etiologies. We used a mouse model of the SCZ-predisposing 22q11.2 deletion (Df(16)A+/- mice) to evaluate how this genetic lesion affects the excitability of layer V prefrontal pyramidal neurons and its modulation by dopamine (DA). Df(16)A+/- mice have normal balance between E/I at baseline but are unable to maintain it upon dopaminergic challenge. Specifically, in wild-type mice, D1 receptor (D1R) activation enhances excitability of layer V prefrontal pyramidal neurons and D2 receptor (D2R) activation reduces it. Whereas the excitatory effect upon D1R activation is enhanced in Df(16)A+/- mice, the inhibitory effect upon D2R activation is reduced. The latter is partly due to the inability of mutant mice to activate GABAergic parvalbumin (PV)+ interneurons through D2Rs. We further demonstrate that reduced KCNQ2 channel function in PV+ interneurons in Df(16)A+/- mice renders them less capable of inhibiting pyramidal neurons upon D2 modulation. Thus, DA modulation of PV+ interneurons and control of E/I are altered in Df(16)A+/- mice with a higher excitation and lower inhibition during dopaminergic modulation.


Assuntos
Dopamina/metabolismo , Interneurônios/fisiologia , Canal de Potássio KCNQ2/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/genética , Esquizofrenia/fisiopatologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Córtex Pré-Frontal/metabolismo , Células Piramidais/fisiologia , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Esquizofrenia/metabolismo
7.
J Neurosci ; 37(15): 4158-4180, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28283561

RESUMO

Using a genetic mouse model that faithfully recapitulates a DISC1 genetic alteration strongly associated with schizophrenia and other psychiatric disorders, we examined the impact of this mutation within the prefrontal cortex. Although cortical layering, cytoarchitecture, and proteome were found to be largely unaffected, electrophysiological examination of the mPFC revealed both neuronal hyperexcitability and alterations in short-term synaptic plasticity consistent with enhanced neurotransmitter release. Increased excitability of layer II/III pyramidal neurons was accompanied by consistent reductions in voltage-activated potassium currents near the action potential threshold as well as by enhanced recruitment of inputs arising from superficial layers to layer V. We further observed reductions in both the paired-pulse ratios and the enhanced short-term depression of layer V synapses arising from superficial layers consistent with enhanced neurotransmitter release at these synapses. Recordings from layer II/III pyramidal neurons revealed action potential widening that could account for enhanced neurotransmitter release. Significantly, we found that reduced functional expression of the voltage-dependent potassium channel subunit Kv1.1 substantially contributes to both the excitability and short-term plasticity alterations that we observed. The underlying dysregulation of Kv1.1 expression was attributable to cAMP elevations in the PFC secondary to reduced phosphodiesterase 4 activity present in Disc1 deficiency and was rescued by pharmacological blockade of adenylate cyclase. Our results demonstrate a potentially devastating impact of Disc1 deficiency on neural circuit function, partly due to Kv1.1 dysregulation that leads to a dual dysfunction consisting of enhanced neuronal excitability and altered short-term synaptic plasticity.SIGNIFICANCE STATEMENT Schizophrenia is a profoundly disabling psychiatric illness with a devastating impact not only upon the afflicted but also upon their families and the broader society. Although the underlying causes of schizophrenia remain poorly understood, a growing body of studies has identified and strongly implicated various specific risk genes in schizophrenia pathogenesis. Here, using a genetic mouse model, we explored the impact of one of the most highly penetrant schizophrenia risk genes, DISC1, upon the medial prefrontal cortex, the region believed to be most prominently dysfunctional in schizophrenia. We found substantial derangements in both neuronal excitability and short-term synaptic plasticity-parameters that critically govern neural circuit information processing-suggesting that similar changes may critically, and more broadly, underlie the neural computational dysfunction prototypical of schizophrenia.


Assuntos
Potenciais de Ação/fisiologia , Modelos Animais de Doenças , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Esquizofrenia/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Córtex Pré-Frontal/fisiopatologia , Gravidez , Esquizofrenia/genética , Esquizofrenia/fisiopatologia
8.
Circulation ; 136(19): 1809-1823, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-28931551

RESUMO

BACKGROUND: Clinical trials of bone marrow cell-based therapies after acute myocardial infarction (MI) have produced mostly neutral results. Treatment with specific bone marrow cell-derived secreted proteins may provide an alternative biological approach to improving tissue repair and heart function after MI. We recently performed a bioinformatic secretome analysis in bone marrow cells from patients with acute MI and discovered a poorly characterized secreted protein, EMC10 (endoplasmic reticulum membrane protein complex subunit 10), showing activity in an angiogenic screen. METHODS: We investigated the angiogenic potential of EMC10 and its mouse homolog (Emc10) in cultured endothelial cells and infarcted heart explants. We defined the cellular sources and function of Emc10 after MI using wild-type, Emc10-deficient, and Emc10 bone marrow-chimeric mice subjected to transient coronary artery ligation. Furthermore, we explored the therapeutic potential of recombinant Emc10 delivered by osmotic minipumps after MI in heart failure-prone FVB/N mice. RESULTS: Emc10 signaled through small GTPases, p21-activated kinase, and the p38 mitogen-activated protein kinase (MAPK)-MAPK-activated protein kinase 2 (MK2) pathway to promote actin polymerization and endothelial cell migration. Confirming the importance of these signaling events in the context of acute MI, Emc10 stimulated endothelial cell outgrowth from infarcted mouse heart explants via p38 MAPK-MK2. Emc10 protein abundance was increased in the infarcted region of the left ventricle and in the circulation of wild-type mice after MI. Emc10 expression was also increased in left ventricular tissue samples from patients with acute MI. Bone marrow-derived monocytes and macrophages were the predominant sources of Emc10 in the infarcted murine heart. Emc10 KO mice showed no cardiovascular phenotype at baseline. After MI, however, capillarization of the infarct border zone was impaired in KO mice, and the animals developed larger infarct scars and more pronounced left ventricular remodeling compared with wild-type mice. Transplanting KO mice with wild-type bone marrow cells rescued the angiogenic defect and ameliorated left ventricular remodeling. Treating FVB/N mice with recombinant Emc10 enhanced infarct border-zone capillarization and exerted a sustained beneficial effect on left ventricular remodeling. CONCLUSIONS: We have identified Emc10 as a previously unknown angiogenic growth factor that is produced by bone marrow-derived monocytes and macrophages as part of an endogenous adaptive response that can be enhanced therapeutically to repair the heart after MI.


Assuntos
Proteínas Angiogênicas/metabolismo , Células da Medula Óssea/metabolismo , Proteínas de Membrana/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Neovascularização Fisiológica , Cicatrização , Proteínas Angiogênicas/administração & dosagem , Proteínas Angiogênicas/deficiência , Proteínas Angiogênicas/genética , Animais , Transplante de Medula Óssea , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Genótipo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana/administração & dosagem , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fatores de Tempo , Cicatrização/efeitos dos fármacos , Quinases Ativadas por p21/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Cereb Cortex ; 26(11): 4265-4281, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27613435

RESUMO

Cognitive deficits represent a major burden of neuropsychiatric disorders and result in part from abnormal communication within hippocampal-prefrontal circuits. While it has been hypothesized that this network dysfunction arises during development, long before the first clinical symptoms, experimental evidence is still missing. Here, we show that pre-juvenile mice mimicking genetic and environmental risk factors of disease (dual-hit GE mice) have poorer recognition memory that correlates with augmented coupling by synchrony and stronger directed interactions between prefrontal cortex and hippocampus. The network dysfunction emerges already during neonatal development, yet it initially consists in a diminished hippocampal theta drive and consequently, a weaker and disorganized entrainment of local prefrontal circuits in discontinuous oscillatory activity in dual-hit GE mice when compared with controls. Thus, impaired maturation of functional communication within hippocampal-prefrontal networks switching from hypo- to hyper-coupling may represent a mechanism underlying the pathophysiology of cognitive deficits in neuropsychiatric disorders.


Assuntos
Transtornos Cognitivos , Deficiências do Desenvolvimento , Interação Gene-Ambiente , Hipocampo/fisiologia , Vias Neurais/fisiologia , Córtex Pré-Frontal/fisiopatologia , Animais , Animais Recém-Nascidos , Transtornos Cognitivos/genética , Transtornos Cognitivos/patologia , Transtornos Cognitivos/fisiopatologia , Deficiências do Desenvolvimento/induzido quimicamente , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/genética , Feminino , Hipocampo/efeitos dos fármacos , Indutores de Interferon/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/efeitos dos fármacos , Poli I-C/toxicidade , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia
10.
Proc Natl Acad Sci U S A ; 111(1): 343-8, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24344280

RESUMO

We used a family-based cluster detection approach designed to localize significant rare disease-risk variants clusters within a region of interest to systematically search for schizophrenia (SCZ) susceptibility genes within 49 genomic loci previously implicated by de novo copy number variants. Using two independent whole-exome sequencing family datasets and a follow-up autism spectrum disorder (ASD) case/control whole-exome sequencing dataset, we identified variants in one gene, Fanconi-associated nuclease 1 (FAN1), as being associated with both SCZ and ASD. FAN1 is located in a region on chromosome 15q13.3 implicated by a recurrent copy number variant, which predisposes to an array of psychiatric and neurodevelopmental phenotypes. In both SCZ and ASD datasets, rare nonsynonymous risk variants cluster significantly in affected individuals within a 20-kb window that spans several key functional domains of the gene. Our finding suggests that FAN1 is a key driver in the 15q13.3 locus for the associated psychiatric and neurodevelopmental phenotypes. FAN1 encodes a DNA repair enzyme, thus implicating abnormalities in DNA repair in the susceptibility to SCZ or ASD.


Assuntos
Transtorno Autístico/genética , Cromossomos Humanos Par 15 , Exodesoxirribonucleases/genética , Predisposição Genética para Doença , Esquizofrenia/genética , Sequência de Aminoácidos , Análise por Conglomerados , Simulação por Computador , Reparo do DNA , Endodesoxirribonucleases , Exoma , Feminino , Marcadores Genéticos , Variação Genética , Humanos , Masculino , Dados de Sequência Molecular , Enzimas Multifuncionais , Nucleotídeos/genética , Fenótipo , Risco , Homologia de Sequência de Aminoácidos , África do Sul , Estados Unidos
11.
Nature ; 464(7289): 763-7, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20360742

RESUMO

Abnormalities in functional connectivity between brain areas have been postulated as an important pathophysiological mechanism underlying schizophrenia. In particular, macroscopic measurements of brain activity in patients suggest that functional connectivity between the frontal and temporal lobes may be altered. However, it remains unclear whether such dysconnectivity relates to the aetiology of the illness, and how it is manifested in the activity of neural circuits. Because schizophrenia has a strong genetic component, animal models of genetic risk factors are likely to aid our understanding of the pathogenesis and pathophysiology of the disease. Here we study Df(16)A(+/-) mice, which model a microdeletion on human chromosome 22 (22q11.2) that constitutes one of the largest known genetic risk factors for schizophrenia. To examine functional connectivity in these mice, we measured the synchronization of neural activity between the hippocampus and the prefrontal cortex during the performance of a task requiring working memory, which is one of the cognitive functions disrupted in the disease. In wild-type mice, hippocampal-prefrontal synchrony increased during working memory performance, consistent with previous reports in rats. Df(16)A(+/-) mice, which are impaired in the acquisition of the task, showed drastically reduced synchrony, measured both by phase-locking of prefrontal cells to hippocampal theta oscillations and by coherence of prefrontal and hippocampal local field potentials. Furthermore, the magnitude of hippocampal-prefrontal coherence at the onset of training could be used to predict the time it took the Df(16)A(+/-) mice to learn the task and increased more slowly during task acquisition. These data suggest how the deficits in functional connectivity observed in patients with schizophrenia may be realized at the single-neuron level. Our findings further suggest that impaired long-range synchrony of neural activity is one consequence of the 22q11.2 deletion and may be a fundamental component of the pathophysiology underlying schizophrenia.


Assuntos
Cromossomos de Mamíferos/genética , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/genética , Esquizofrenia/fisiopatologia , Potenciais de Ação/fisiologia , Alelos , Animais , Comportamento Animal/fisiologia , Cromossomos Humanos Par 22/genética , Feminino , Predisposição Genética para Doença/genética , Humanos , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Modelos Neurológicos
12.
Neurobiol Dis ; 77: 228-37, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25771167

RESUMO

Variation in gene expression is an important mechanism underlying susceptibility to complex disease and traits. Single nucleotide polymorphisms (SNPs) account for a substantial portion of the total detected genetic variation in gene expression but how exactly variants acting in trans modulate gene expression and disease susceptibility remains largely unknown. The BDNF Val66Met SNP has been associated with a number of psychiatric disorders such as depression, anxiety disorders, schizophrenia and related traits. Using global microRNA expression profiling in hippocampus of humanized BDNF Val66Met knock-in mice we showed that this variant results in dysregulation of at least one microRNA, which in turn affects downstream target genes. Specifically, we show that reduced levels of miR-146b (mir146b), lead to increased Per1 and Npas4 mRNA levels and increased Irak1 protein levels in vitro and are associated with similar changes in the hippocampus of hBDNF(Met/Met) mice. Our findings highlight trans effects of common variants on microRNA-mediated gene expression as an integral part of the genetic architecture of complex disorders and traits.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Regulação da Expressão Gênica/genética , Metionina/genética , MicroRNAs/metabolismo , Valina/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas Circadianas Period/metabolismo
13.
Nat Rev Neurosci ; 11(6): 402-16, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20485365

RESUMO

Recent studies are beginning to paint a clear and consistent picture of the impairments in psychological and cognitive competencies that are associated with microdeletions in chromosome 22q11.2. These studies have highlighted a strong link between this genetic lesion and schizophrenia. Parallel studies in humans and animal models are starting to uncover the complex genetic and neural substrates altered by the microdeletion. In addition to offering a deeper understanding of the effects of this genetic lesion, these findings may guide analysis of other copy-number variants associated with cognitive dysfunction and psychiatric disorders.


Assuntos
Encefalopatias/etiologia , Aberrações Cromossômicas , Deleção Cromossômica , Cromossomos Humanos Par 22 , Esquizofrenia , Animais , Encefalopatias/genética , Humanos , Esquizofrenia/complicações , Esquizofrenia/genética , Esquizofrenia/patologia
14.
J Neurosci ; 33(37): 14825-39, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24027283

RESUMO

We used a mouse model of the schizophrenia-predisposing 22q11.2 microdeletion to evaluate how this genetic lesion affects cortical neural circuits at the synaptic, cellular, and molecular levels. Guided by cognitive deficits, we demonstrated that mutant mice display robust deficits in high-frequency synaptic transmission and short-term plasticity (synaptic depression and potentiation), as well as alterations in long-term plasticity and dendritic spine stability. Apart from previously reported reduction in dendritic complexity of layer 5 pyramidal neurons, altered synaptic plasticity occurs in the context of relatively circumscribed and often subtle cytoarchitectural changes in neuronal density and inhibitory neuron numbers. We confirmed the pronounced DiGeorge critical region 8 (Dgcr8)-dependent deficits in primary micro-RNA processing and identified additional changes in gene expression and RNA splicing that may underlie the effects of this mutation. Reduction in Dgcr8 levels appears to be a major driver of altered short-term synaptic plasticity in prefrontal cortex and working memory but not of long-term plasticity and cytoarchitecture. Our findings inform the cortical synaptic and neuronal mechanisms of working memory impairment in the context of psychiatric disorders. They also provide insight into the link between micro-RNA dysregulation and genetic liability to schizophrenia and cognitive dysfunction.


Assuntos
Síndrome de DiGeorge/patologia , Potenciação de Longa Duração/genética , Depressão Sináptica de Longo Prazo/genética , Neurônios/fisiologia , Córtex Pré-Frontal/patologia , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/genética , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Síndrome de DiGeorge/complicações , Síndrome de DiGeorge/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Fosfopiruvato Hidratase/metabolismo , Proteínas/genética , Proteínas de Ligação a RNA , Reconhecimento Psicológico/fisiologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo
15.
Annu Rev Med ; 63: 63-80, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22034867

RESUMO

Although a genetic component of schizophrenia has been acknowledged for a long time, the underlying architecture of the genetic risk remains a contentious issue. Early linkage and candidate association studies led to largely inconclusive results. More recently, the availability of powerful technologies, samples of sufficient sizes, and genome-wide panels of genetic markers facilitated systematic and agnostic scans throughout the genome for either common or rare disease risk variants of small or large effect size, respectively. Although the former had limited success, the role of rare genetic events, such as copy-number variants (CNVs) or rare point mutations, has become increasingly important in gene discovery for schizophrenia. Importantly, recent research building upon earlier findings of de novo recurrent CNVs at the 22q11.2 locus, has highlighted a de novo mutational paradigm as a major component of the genetic architecture of schizophrenia. Recent progress is bringing us closer to earlier intervention and new therapeutic targets.


Assuntos
Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Genética/tendências , Esquizofrenia/epidemiologia , Esquizofrenia/genética , Humanos , Fatores de Risco
16.
Proc Natl Acad Sci U S A ; 108(11): 4447-52, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21368174

RESUMO

Individuals with 22q11.2 microdeletions have cognitive and behavioral impairments and the highest known genetic risk for developing schizophrenia. One gene disrupted by the 22q11.2 microdeletion is DGCR8, a component of the "microprocessor" complex that is essential for microRNA production, resulting in abnormal processing of specific brain miRNAs and working memory deficits. Here, we determine the effect of Dgcr8 deficiency on the structure and function of cortical circuits by assessing their laminar organization, as well as the neuronal morphology, and intrinsic and synaptic properties of layer 5 pyramidal neurons in the prefrontal cortex of Dgcr8(+/-) mutant mice. We found that heterozygous Dgcr8 mutant mice have slightly fewer cortical layer 2/4 neurons and that the basal dendrites of layer 5 pyramidal neurons have slightly smaller spines. In addition to the modest structural changes, field potential and whole-cell electrophysiological recordings performed in layer 5 of the prefrontal cortex revealed greater short-term synaptic depression during brief stimulation trains applied at 50 Hz to superficial cortical layers. This finding was accompanied by a decrease in the initial phase of synaptic potentiation. Our results identify altered short-term plasticity as a neural substrate underlying the cognitive dysfunction and the increased risk for schizophrenia associated with the 22q11.2 microdeletions.


Assuntos
Deleção de Genes , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal/fisiopatologia , Proteínas/metabolismo , Animais , Região CA1 Hipocampal/fisiopatologia , Região CA3 Hipocampal/fisiopatologia , Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Camundongos , Córtex Pré-Frontal/patologia , Proteínas de Ligação a RNA , Sinapses/metabolismo , Fatores de Tempo
17.
Proc Natl Acad Sci U S A ; 108(49): E1349-58, 2011 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22049344

RESUMO

Carefully designed animal models of genetic risk factors are likely to aid our understanding of the pathogenesis of schizophrenia. Here, we study a mouse strain with a truncating lesion in the endogenous Disc1 ortholog designed to model the effects of a schizophrenia-predisposing mutation and offer a detailed account of the consequences that this mutation has on the development and function of a hippocampal circuit. We uncover widespread and cumulative cytoarchitectural alterations in the dentate gyrus during neonatal and adult neurogenesis, which include errors in axonal targeting and are accompanied by changes in short-term plasticity at the mossy fiber/CA3 circuit. We also provide evidence that cAMP levels are elevated as a result of the Disc1 mutation, leading to altered axonal targeting and dendritic growth. The identified structural alterations are, for the most part, not consistent with the growth-promoting and premature maturation effects inferred from previous RNAi-based Disc1 knockdown. Our results provide support to the notion that modest disturbances of neuronal connectivity and accompanying deficits in short-term synaptic dynamics is a general feature of schizophrenia-predisposing mutations.


Assuntos
Axônios/metabolismo , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal , Potenciais de Ação , Animais , Animais Recém-Nascidos , Proliferação de Células , Células Cultivadas , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Dendritos/metabolismo , Dendritos/fisiologia , Giro Denteado/citologia , Giro Denteado/crescimento & desenvolvimento , Giro Denteado/metabolismo , Hipocampo/citologia , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Potenciação de Longa Duração , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fibras Musgosas Hipocampais/metabolismo , Proteínas do Tecido Nervoso/genética , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Neurônios/fisiologia , Técnicas de Patch-Clamp
18.
Cancer Discov ; 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38767413

RESUMO

High-grade gliomas (HGG) are deadly diseases for both adult and pediatric patients. Recently, it has been shown that neuronal activity promotes progression of multiple subgroups of HGG. However, epigenetic mechanisms that govern this process remain elusive. Here we report that the chromatin remodeler CHD2 regulates neuron-glioma interactions in diffuse midline glioma (DMG) characterized by onco-histone H3.1K27M. Depletion of CHD2 in H3.1K27M DMG cells compromises cell viability and neuron-to-glioma synaptic connections in vitro, neuron-induced proliferation of H3.1K27M DMG cells in vitro and in vivo, activity-dependent calcium transients in vivo, and extends the survival of H3.1K27M DMG-bearing mice. Mechanistically, CHD2 coordinates with the transcription factor FOSL1 to control the expression of axon-guidance and synaptic genes in H3.1K27M DMG cells. Together, our study reveals a mechanism whereby CHD2 controls the intrinsic gene program of the H3.1K27M DMG subtype, which in turn regulates the tumor growth-promoting interactions of glioma cells with neurons.

19.
Sci Am ; 319(4): 10, 2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30273301
20.
Nat Genet ; 36(2): 131-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14745448

RESUMO

AKT-GSK3beta signaling is a target of lithium and as such has been implicated in the pathogenesis of mood disorders. Here, we provide evidence that this signaling pathway also has a role in schizophrenia. Specifically, we present convergent evidence for a decrease in AKT1 protein levels and levels of phosphorylation of GSK3beta at Ser9 in the peripheral lymphocytes and brains of individuals with schizophrenia; a significant association between schizophrenia and an AKT1 haplotype associated with lower AKT1 protein levels; and a greater sensitivity to the sensorimotor gating-disruptive effect of amphetamine, conferred by AKT1 deficiency. Our findings support the proposal that alterations in AKT1-GSK3beta signaling contribute to schizophrenia pathogenesis and identify AKT1 as a potential schizophrenia susceptibility gene. Consistent with this proposal, we also show that haloperidol induces a stepwise increase in regulatory phosphorylation of AKT1 in the brains of treated mice that could compensate for an impaired function of this signaling pathway in schizophrenia.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Esquizofrenia/enzimologia , Transdução de Sinais/fisiologia , Antipsicóticos/farmacologia , Glicogênio Sintase Quinase 3 beta , Haloperidol/farmacologia , Haplótipos , Humanos , Fosforilação/efeitos dos fármacos , Fosfotransferases/efeitos dos fármacos , Fosfotransferases/metabolismo , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt , Esquizofrenia/genética , Esquizofrenia/metabolismo , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa