Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Neurobiol Dis ; 127: 210-222, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30831192

RESUMO

Autism spectrum disorders are early onset neurodevelopmental disorders characterized by deficits in social communication and restricted repetitive behaviors, yet they are quite heterogeneous in terms of their genetic basis and phenotypic manifestations. Recently, de novo pathogenic mutations in DYRK1A, a chromosome 21 gene associated to neuropathological traits of Down syndrome, have been identified in patients presenting a recognizable syndrome included in the autism spectrum. These mutations produce DYRK1A kinases with partial or complete absence of the catalytic domain, or they represent missense mutations located within this domain. Here, we undertook an extensive biochemical characterization of the DYRK1A missense mutations reported to date and show that most of them, but not all, result in enzymatically dead DYRK1A proteins. We also show that haploinsufficient Dyrk1a+/- mutant mice mirror the neurological traits associated with the human pathology, such as defective social interactions, stereotypic behaviors and epileptic activity. These mutant mice present altered proportions of excitatory and inhibitory neocortical neurons and synapses. Moreover, we provide evidence that alterations in the production of cortical excitatory neurons are contributing to these defects. Indeed, by the end of the neurogenic period, the expression of developmental regulated genes involved in neuron differentiation and/or activity is altered. Therefore, our data indicate that altered neocortical neurogenesis could critically affect the formation of cortical circuits, thereby contributing to the neuropathological changes in DYRK1A haploinsufficiency syndrome.


Assuntos
Transtorno Autístico/metabolismo , Haploinsuficiência , Neocórtex/metabolismo , Rede Nervosa/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Comportamento Social , Animais , Transtorno Autístico/genética , Comportamento Animal/fisiologia , Masculino , Camundongos , Mutação de Sentido Incorreto , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Quinases Dyrk
2.
PLoS Genet ; 11(3): e1005062, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25803843

RESUMO

The trisomy of human chromosome 21 (Hsa21), which causes Down syndrome (DS), is the most common viable human aneuploidy. In contrast to trisomy, the complete monosomy (M21) of Hsa21 is lethal, and only partial monosomy or mosaic monosomy of Hsa21 is seen. Both conditions lead to variable physiological abnormalities with constant intellectual disability, locomotor deficits, and altered muscle tone. To search for dosage-sensitive genes involved in DS and M21 phenotypes, we created two new mouse models: the Ts3Yah carrying a tandem duplication and the Ms3Yah carrying a deletion of the Hspa13-App interval syntenic with 21q11.2-q21.3. Here we report that the trisomy and the monosomy of this region alter locomotion, muscle strength, mass, and energetic balance. The expression profiling of skeletal muscles revealed global changes in the regulation of genes implicated in energetic metabolism, mitochondrial activity, and biogenesis. These genes are downregulated in Ts3Yah mice and upregulated in Ms3Yah mice. The shift in skeletal muscle metabolism correlates with a change in mitochondrial proliferation without an alteration in the respiratory function. However, the reactive oxygen species (ROS) production from mitochondrial complex I decreased in Ms3Yah mice, while the membrane permeability of Ts3Yah mitochondria slightly increased. Thus, we demonstrated how the Hspa13-App interval controls metabolic and mitochondrial phenotypes in muscles certainly as a consequence of change in dose of Gabpa, Nrip1, and Atp5j. Our results indicate that the copy number variation in the Hspa13-App region has a peripheral impact on locomotor activity by altering muscle function.


Assuntos
Síndrome de Down/genética , Monossomia/genética , Atividade Motora/genética , Força Muscular/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cromossomos Humanos Par 21/genética , Modelos Animais de Doenças , Síndrome de Down/fisiopatologia , Metabolismo Energético/genética , Fator de Transcrição de Proteínas de Ligação GA/genética , Humanos , Camundongos , Mitocôndrias Musculares/genética , Mitocôndrias Musculares/patologia , ATPases Mitocondriais Próton-Translocadoras/genética , Monossomia/fisiopatologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Proteínas Nucleares/genética , Proteína 1 de Interação com Receptor Nuclear
3.
Hum Mol Genet ; 23(24): 6481-94, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027326

RESUMO

Williams-Beuren syndrome is a developmental multisystemic disorder caused by a recurrent 1.55-1.83 Mb heterozygous deletion on human chromosome band 7q11.23. Through chromosomal engineering with the cre-loxP system, we have generated mice with an almost complete deletion (CD) of the conserved syntenic region on chromosome 5G2. Heterozygous CD mice were viable, fertile and had a normal lifespan, while homozygotes were early embryonic lethal. Transcript levels of most deleted genes were reduced 50% in several tissues, consistent with gene dosage. Heterozygous mutant mice showed postnatal growth delay with reduced body weight and craniofacial abnormalities such as small mandible. The cardiovascular phenotype was only manifested with borderline hypertension, mildly increased arterial wall thickness and cardiac hypertrophy. The neurobehavioral phenotype revealed impairments in motor coordination, increased startle response to acoustic stimuli and hypersociability. Mutant mice showed a general reduction in brain weight. Cellular and histological abnormalities were present in the amygdala, cortex and hippocampus, including increased proportion of immature neurons. In summary, these mice recapitulate most crucial phenotypes of the human disorder, provide novel insights into the pathophysiological mechanisms of the disease such as the neural substrates of the behavioral manifestations, and will be valuable to evaluate novel therapeutic approaches.


Assuntos
Deleção Cromossômica , Cromossomos de Mamíferos , Anormalidades Craniofaciais/genética , Modelos Animais de Doenças , Síndrome de Williams/genética , Animais , Comportamento Animal , Peso Corporal , Encéfalo/patologia , Encéfalo/fisiopatologia , Cardiomegalia/fisiopatologia , Cromossomos Humanos Par 7 , Anormalidades Craniofaciais/patologia , Anormalidades Craniofaciais/fisiopatologia , Feminino , Dosagem de Genes , Genes Letais , Heterozigoto , Humanos , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Neurônios/patologia , Tamanho do Órgão , Sintenia , Síndrome de Williams/patologia , Síndrome de Williams/fisiopatologia
4.
Mol Ther ; 23(11): 1691-1699, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26216516

RESUMO

Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder caused by a heterozygous deletion of 26-28 genes at chromosome band 7q11.23. Haploinsufficiency at GTF2I has been shown to play a major role in the neurobehavioral phenotype. By characterizing the neuronal architecture in four animal models with intragenic, partial, and complete deletions of the WBS critical interval (ΔGtf2i(+/-), ΔGtf2i( -/-), PD, and CD), we clarify the involvement of Gtf2i in neurocognitive features. All mutant mice showed hypersociability, impaired motor learning and coordination, and altered anxiety-like behavior. Dendritic length was decreased in the CA1 of ΔGtf2i(+/-), ΔGtf2i ( -/-), and CD mice. Spine density was reduced, and spines were shorter in ΔGtf2i ( -/-), PD, and CD mice. Overexpression of Pik3r1 and downregulation of Bdnf were observed in ΔGtf2i(+/-), PD, and CD mice. Intracisternal Gtf2i-gene therapy in CD mice using adeno-associated virus resulted in increased mGtf2i expression and normalization of Bdnf levels, along with beneficial effects in motor coordination, sociability, and anxiety, despite no significant changes in neuronal architecture. Our findings further indicate that Gtf2i haploinsufficiency plays an important role in the neurodevelopmental and cognitive abnormalities of WBS and that it is possible to rescue part of this neurocognitive phenotype by restoring Gtf2i expression levels in specific brain areas.


Assuntos
Cisterna Magna , Cognição , Plasticidade Neuronal/genética , Fatores de Transcrição TFII/genética , Síndrome de Williams/terapia , Animais , Comportamento Animal , Cromossomos Humanos Par 7/genética , Dependovirus , Modelos Animais de Doenças , Deleção de Genes , Terapia Genética , Haploinsuficiência , Humanos , Injeções Espinhais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fatores de Transcrição TFII/fisiologia , Síndrome de Williams/fisiopatologia
5.
Neurobiol Dis ; 63: 92-106, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24291518

RESUMO

PCP4/PEP19 is a modulator of Ca(2+)-CaM signaling. In the brain, it is expressed in a very specific pattern in postmitotic neurons. In particular, Pcp4 is highly expressed in the Purkinje cell, the sole output neuron of the cerebellum. PCP4, located on human chromosome 21, is present in three copies in individuals with Down syndrome (DS). In a previous study using a transgenic mouse model (TgPCP4) to evaluate the consequences of 3 copies of this gene, we found that PCP4 overexpression induces precocious neuronal differentiation during mouse embryogenesis. Here, we report combined analyses of the cerebellum at postnatal stages (P14 and adult) in which we identified age-related molecular, electrophysiological, and behavioral alterations in the TgPCP4 mouse. While Pcp4 overexpression at P14 induces an earlier neuronal maturation, at adult stage it induces increase in cerebellar CaMK2alpha and in cerebellar LTD, as well as learning impairments. We therefore propose that PCP4 contributes significantly to the development of Down syndrome phenotypes through molecular and functional changes.


Assuntos
Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Recém-Nascidos , Cerebelo/citologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Humanos , Técnicas In Vitro , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/genética , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Quinoxalinas/farmacologia , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrodotoxina/farmacologia , Valina/análogos & derivados , Valina/farmacologia
6.
Neurobiol Dis ; 69: 65-75, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24801365

RESUMO

Cognitive deficits in Down syndrome (DS) have been linked to increased synaptic inhibition, leading to an imbalance of excitation/inhibition (E/I). Various mouse models and studies from human brains have implicated an HSA21 gene, the serine/threonine kinase DYRK1A, as a candidate for inducing cognitive dysfunction. Here, consequences of alterations in Dyrk1a dosage were assessed in mouse models with varying copy numbers of Dyrk1a: mBACtgDyrk1a, Ts65Dn and Dp(16)1Yey (with 3 gene copies) and Dyrk1a(+/-) (one functional copy). Molecular (i.e. immunoblotting/immunohistochemistry) and behavioral analyses (e.g., rotarod, Morris water maze, Y-maze) were performed in mBACtgDyrk1a mice. Increased expression of DYRK1A in mBACtgDyrk1a induced molecular alterations in synaptic plasticity pathways, particularly expression changes in GABAergic and glutaminergic related proteins. Similar alterations were observed in models with partial trisomy of MMU16, Ts65Dn and Dp(16)1Yey, and were reversed in the Dyrk1a(+/-) model. Dyrk1a overexpression produced an increased number and signal intensity of GAD67 positive neurons, indicating enhanced inhibition pathways in three different models: mBACtgDyrk1a, hYACtgDyrk1a and Dp(16)1Yey. Functionally, Dyrk1a overexpression protected mice from PTZ-induced seizures related to GABAergic neuron plasticity. Our study shows that DYRK1A overexpression affects pathways involved in synaptogenesis and synaptic plasticity and influences E/I balance toward inhibition. Inhibition of DYRK1A activity offers a therapeutic target for DS, but its inhibition/activation may also be relevant for other psychiatric diseases with E/I balance alterations.


Assuntos
Dosagem de Genes , Aprendizagem , Inibição Neural/genética , Plasticidade Neuronal/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Animais , Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/fisiopatologia , Síndrome de Down/psicologia , Humanos , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/genética , Atividade Motora/fisiologia , Inibição Neural/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Convulsões/genética , Convulsões/fisiopatologia , Sinapses/genética , Sinapses/fisiologia , Quinases Dyrk
7.
J Biol Chem ; 287(25): 21224-32, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22539351

RESUMO

The brain-specific isoform carnitine palmitoyltransferase 1C (CPT1C) has been implicated in the hypothalamic regulation of food intake and energy homeostasis. Nevertheless, its molecular function is not completely understood, and its role in other brain areas is unknown. We demonstrate that CPT1C is expressed in pyramidal neurons of the hippocampus and is located in the endoplasmic reticulum throughout the neuron, even inside dendritic spines. We used molecular, cellular, and behavioral approaches to determine CPT1C function. First, we analyzed the implication of CPT1C in ceramide metabolism. CPT1C overexpression in primary hippocampal cultured neurons increased ceramide levels, whereas in CPT1C-deficient neurons, ceramide levels were diminished. Correspondingly, CPT1C knock-out (KO) mice showed reduced ceramide levels in the hippocampus. At the cellular level, CPT1C deficiency altered dendritic spine morphology by increasing immature filopodia and reducing mature mushroom and stubby spines. Total protrusion density and spine head area in mature spines were unaffected. Treatment of cultured neurons with exogenous ceramide reverted the KO phenotype, as did ectopic overexpression of CPT1C, indicating that CPT1C regulation of spine maturation is mediated by ceramide. To study the repercussions of the KO phenotype on cognition, we performed the hippocampus-dependent Morris water maze test on mice. Results show that CPT1C deficiency strongly impairs spatial learning. All of these results demonstrate that CPT1C regulates the levels of ceramide in the endoplasmic reticulum of hippocampal neurons, and this is a relevant mechanism for the correct maturation of dendritic spines and for proper spatial learning.


Assuntos
Carnitina O-Palmitoiltransferase/biossíntese , Ceramidas/metabolismo , Dendritos/enzimologia , Metabolismo Energético/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Metabolismo dos Lipídeos/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Células Piramidais/enzimologia , Animais , Comportamento Animal/fisiologia , Carnitina O-Palmitoiltransferase/genética , Células Cultivadas , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Erros Inatos do Metabolismo Lipídico/enzimologia , Erros Inatos do Metabolismo Lipídico/genética , Erros Inatos do Metabolismo Lipídico/patologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Células Piramidais/citologia
8.
J Neurochem ; 124(3): 347-62, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23216354

RESUMO

The mitochondrial transporter of aspartate-glutamate Aralar/AGC1 is a regulatory component of the malate-aspartate shuttle. Aralar deficiency in mouse and human causes a shutdown of brain shuttle activity and global cerebral hypomyelination. A lack of neurofilament-labeled processes is detected in the cerebral cortex, but whether different types of neurons are differentially affected by Aralar deficiency is still unknown. We have now found that Aralar-knockout (Aralar-KO) post-natal mice show hyperactivity, anxiety-like behavior, and hyperreactivity with a decrease of dopamine (DA) in terminal-rich regions. The striatum is the brain region most affected in terms of size, amino acid and monoamine content. We find a decline in vesicular monoamine transporter-2 (VMAT2) levels associated with increased DA metabolism through MAO activity (DOPAC/DA ratio) in Aralar-KO striatum. However, no decrease in DA or in the number of nigral tyrosine hydroxylase-positive cells was detected in Aralar-KO brainstem. Adult Aralar-hemizygous mice presented also increased DOPAC/DA ratio in striatum and enhanced sensitivity to amphetamine. Our results suggest that Aralar deficiency causes a fall in GSH/GSSG ratio and VMAT2 in striatum that might be related to a failure to produce mitochondrial NADH and to an increase of reactive oxygen species (ROS) in the cytosol. The results indicate that the nigrostriatal dopaminergic system is a target of Aralar deficiency.


Assuntos
Ácido Aspártico/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Malatos/metabolismo , Doenças Mitocondriais/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transtornos Psicomotores/metabolismo , Substância Negra/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/deficiência , Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Antiporters/deficiência , Antiporters/genética , Antiporters/metabolismo , Ácido Aspártico/fisiologia , Corpo Estriado/citologia , Dopamina/deficiência , Dopamina/genética , Emoções/fisiologia , Comportamento Exploratório/fisiologia , Feminino , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças Mitocondriais/genética , Doenças Mitocondriais/fisiopatologia , Proteínas de Transporte da Membrana Mitocondrial/deficiência , Proteínas de Transporte da Membrana Mitocondrial/genética , Transtornos das Habilidades Motoras/genética , Transtornos das Habilidades Motoras/metabolismo , Vias Neurais/citologia , Vias Neurais/metabolismo , Vias Neurais/fisiopatologia , Estresse Oxidativo/fisiologia , Gravidez , Transtornos Psicomotores/genética , Transtornos Psicomotores/fisiopatologia , Substância Negra/citologia
9.
Hum Mol Genet ; 18(24): 4756-69, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19783846

RESUMO

Mental retardation in Down syndrome (DS), the most frequent trisomy in humans, varies from moderate to severe. Several studies both in human and based on mouse models identified some regions of human chromosome 21 (Hsa21) as linked to cognitive deficits. However, other intervals such as the telomeric region of Hsa21 may contribute to the DS phenotype but their role has not yet been investigated in detail. Here we show that the trisomy of the 12 genes, found in the 0.59 Mb (Abcg1-U2af1) Hsa21 sub-telomeric region, in mice (Ts1Yah) produced defects in novel object recognition, open-field and Y-maze tests, similar to other DS models, but induces an improvement of the hippocampal-dependent spatial memory in the Morris water maze along with enhanced and longer lasting long-term potentiation in vivo in the hippocampus. Overall, we demonstrate the contribution of the Abcg1-U2af1 genetic region to cognitive defect in working and short-term recognition memory in DS models. Increase in copy number of the Abcg1-U2af1 interval leads to an unexpected gain of cognitive function in spatial learning. Expression analysis pinpoints several genes, such as Ndufv3, Wdr4, Pknox1 and Cbs, as candidates whose overexpression in the hippocampus might facilitate learning and memory in Ts1Yah mice. Our work unravels the complexity of combinatorial genetic code modulating different aspect of mental retardation in DS patients. It establishes definitely the contribution of the Abcg1-U2af1 orthologous region to the DS etiology and suggests new modulatory pathways for learning and memory.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Modelos Animais de Doenças , Síndrome de Down/genética , Lipoproteínas/genética , Camundongos , Proteínas Nucleares/genética , Ribonucleoproteínas/genética , Trissomia/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Ansiedade/genética , Sinapses Elétricas/fisiologia , Comportamento Exploratório , Deleção de Genes , Dosagem de Genes , Duplicação Gênica , Código Genético , Humanos , Aprendizagem , Memória , Camundongos Mutantes , Atividade Motora/genética , Fator de Processamento U2AF
10.
Am J Hum Genet ; 83(4): 479-88, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18940310

RESUMO

Genetic-dissection studies carried out with Down syndrome (DS) murine models point to the critical contribution of Dyrk1A overexpression to the motor abnormalities and cognitive deficits displayed in DS individuals. In the present study we have used a murine model overexpressing Dyrk1A (TgDyrk1A mice) to evaluate whether functional CNS defects could be corrected with an inhibitory RNA against Dyrk1A, delivered by bilateral intrastriatal injections of adeno-associated virus type 2 (AAVshDyrk1A). We report that AAVshDyrk1A efficiently transduced HEK293 cells and primary neuronal cultures, triggering the specific inhibition of Dyrk1A expression. Injecting the vector into the striata of TgDyrk1A mice resulted in a restricted, long-term transduction of the striatum. This gene therapy was found to be devoid of toxicity and succeeded in normalizing Dyrk1A protein levels in TgDyrk1A mice. Importantly, the behavioral studies of the adult TgDyrk1A mice treated showed a reversal of corticostriatal-dependent phenotypes, as revealed by the attenuation of their hyperactive behavior, the restoration of motor-coordination defects, and an improvement in sensorimotor gating. Taken together, the data demonstrate that normalizing Dyrk1A gene expression in the striatum of adult TgDyrk1A mice, by means of AAVshRNA, clearly reverses motor impairment. Furthermore, these results identify Dyrk1A as a potential target for therapy in DS.


Assuntos
Síndrome de Down/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Animais , Comportamento Animal , Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Dependovirus/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Modelos Genéticos , Neurônios/metabolismo , Ratos , Quinases Dyrk
11.
Addict Biol ; 14(4): 373-83, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19740365

RESUMO

The increase in the incidence of obesity and eating disorders has promoted research aimed at understanding the aetiology of abnormal eating behaviours. Apart from metabolic factors, obesity is caused by overeating. Clinical reports have led to the suggestion that some individuals may develop addictive-like behaviours when consuming palatable foods, and compulsive eating plays a similar dominant role in obesity as compulsive drug taking does in drug addiction. The progress made in the development of treatment strategies for obesity is limited, in part, because the physiological and neurological causes and consequences of compulsive eating behaviour are not clearly understood and cannot readily be studied in human subjects. We have developed experimental approaches that reflect the functioning of the components of eating control, including compulsive food taking in rats. Rats that are given free choice between standard chow and a palatable, chocolate-containing 'Cafeteria Diet' (CD) develop distinct signs of compulsive food taking that appear at an early stage. These include the inability to adapt intake behaviour in periods of limited or bitter-tasting CD access, continued food intake during resting phases and changes in fine structure of feeding (duration, distribution and recurrence of feeding bouts). The model will help examine the neurobiological underpinnings of compulsive food seeking and food taking and provides a possibility to study the effects of novel anti-obesity compounds on compulsive eating and other components of food-taking behaviour in detail. For future use of genetic models, the possibility of a transfer to a mouse was discussed.


Assuntos
Comportamento Compulsivo , Comportamento Alimentar , Animais , Transtorno da Compulsão Alimentar/epidemiologia , Peso Corporal , Comportamento de Escolha , Modelos Animais de Doenças , Etanol , Feminino , Obesidade/epidemiologia , Ratos , Ratos Wistar , Recompensa , Sacarose , Paladar
12.
J Neurosci ; 27(9): 2253-60, 2007 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17329422

RESUMO

Neurotrophins and their cognate receptors might serve as feedback regulators for the efficacy of synaptic transmission. We analyzed mice overexpressing TrkC (TgNTRK3) for synaptic plasticity and the expression of glutamate receptor subunits. Animals were conditioned using a trace [conditioned stimulus (CS), tone; unconditioned stimulus (US), shock] paradigm. A single electrical pulse presented to the Schaffer collateral-commissural pathway during the CS-US interval evoked a monosynaptic field EPSP (fEPSP) at ipsilateral CA1 pyramidal cells. In wild types, fEPSP slopes increased across conditioning sessions and decreased during extinction, being linearly related to learning evolution. In contrast, fEPSPs in TgNTRK3 animals reached extremely high values, not accompanied with a proportionate increase in their learning curves. Long-term potentiation evoked in conscious TgNTRK3 was also significantly longer lasting than in wild-type mice. These functional alterations were accompanied by significant changes in NR1 and NR2B NMDA receptor subunits, with no modification of NR1(Ser 896) or NR1(Ser 897) phosphorylation. No changes of AMPA and kainate subunits were detected. Results indicate that the NT-3/TrkC cascade could regulate synaptic transmission and plasticity through modulation of glutamatergic transmission at the CA3-CA1 synapse.


Assuntos
Aprendizagem por Associação/fisiologia , Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Condicionamento Clássico , Potenciais Evocados , Hipocampo/citologia , Potenciação de Longa Duração/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/genética , N-Metilaspartato/metabolismo , Fatores de Crescimento Neural/metabolismo , Receptor trkC/genética , Receptor trkC/metabolismo
13.
Elife ; 72018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29355479

RESUMO

Age-related hearing loss (ARHL) is the most common sensory deficit in the elderly. The disease has a multifactorial etiology with both environmental and genetic factors involved being largely unknown. SLC7A8/SLC3A2 heterodimer is a neutral amino acid exchanger. Here, we demonstrated that SLC7A8 is expressed in the mouse inner ear and that its ablation resulted in ARHL, due to the damage of different cochlear structures. These findings make SLC7A8 transporter a strong candidate for ARHL in humans. Thus, a screening of a cohort of ARHL patients and controls was carried out revealing several variants in SLC7A8, whose role was further investigated by in vitro functional studies. Significant decreases in SLC7A8 transport activity was detected for patient's variants (p.Val302Ile, p.Arg418His, p.Thr402Met and p.Val460Glu) further supporting a causative role for SLC7A8 in ARHL. Moreover, our preliminary data suggest that a relevant proportion of ARHL cases could be explained by SLC7A8 mutations.


Assuntos
Mutação , Presbiacusia/genética , Presbiacusia/patologia , Sistema y+ de Transporte de Aminoácidos/deficiência , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Cadeias Leves da Proteína-1 Reguladora de Fusão/deficiência , Cadeias Leves da Proteína-1 Reguladora de Fusão/genética , Deleção de Genes , Testes Genéticos , Humanos , Camundongos
14.
Genetics ; 197(3): 899-912, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24752061

RESUMO

Down syndrome (DS) is due to increased copy number of human chromosome 21. The contribution of different genetic regions has been tested using mouse models. As shown previously, the Abcg1-U2af1 genetic region contributes to cognitive defects in working and short-term recognition memory in Down syndrome mouse models. Here we analyzed the impact of monosomy of the same genetic interval, using a new mouse model, named Ms2Yah. We used several cognitive paradigms and did not detect defects in the object recognition or the Morris water maze tests. However, surprisingly, Ms2Yah mice displayed increased associative memory in a pure contextual fear-conditioning test and decreased social novelty interaction along with a larger long-term potentiation recorded in the CA1 area following stimulation of Schaffer collaterals. Whole-genome expression studies carried out on hippocampus showed that the transcription of only a small number of genes is affected, mainly from the genetic interval (Cbs, Rsph1, Wdr4), with a few additional ones, including the postsynaptic Gabrr2, Gabbr1, Grid2p, Park2, and Dlg1 and the components of the Ubiquitin-mediated proteolysis (Anapc1, Rnf7, Huwe1, Park2). The Abcg1-U2af1 region is undeniably encompassing dosage-sensitive genes or elements whose change in copy number directly affects learning and memory, synaptic function, and autistic related behavior.


Assuntos
Cognição , Síndrome de Down/genética , Síndrome de Down/fisiopatologia , Genoma , Hipocampo/fisiopatologia , Monossomia/genética , Plasticidade Neuronal , Animais , Comportamento Animal , Análise por Conglomerados , Condicionamento Psicológico , Modelos Animais de Doenças , Comportamento Exploratório , Regulação da Expressão Gênica , Hipocampo/patologia , Aprendizagem , Potenciação de Longa Duração , Camundongos Endogâmicos C57BL , Monossomia/fisiopatologia , Atividade Motora , Comportamento Social
15.
Mol Cell Biol ; 34(5): 877-87, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24366545

RESUMO

Changes in nuclear Ca(2+) homeostasis activate specific gene expression programs and are central to the acquisition and storage of information in the brain. DREAM (downstream regulatory element antagonist modulator), also known as calsenilin/KChIP-3 (K(+) channel interacting protein 3), is a Ca(2+)-binding protein that binds DNA and represses transcription in a Ca(2+)-dependent manner. To study the function of DREAM in the brain, we used transgenic mice expressing a Ca(2+)-insensitive/CREB-independent dominant active mutant DREAM (daDREAM). Using genome-wide analysis, we show that DREAM regulates the expression of specific activity-dependent transcription factors in the hippocampus, including Npas4, Nr4a1, Mef2c, JunB, and c-Fos. Furthermore, DREAM regulates its own expression, establishing an autoinhibitory feedback loop to terminate activity-dependent transcription. Ablation of DREAM does not modify activity-dependent transcription because of gene compensation by the other KChIP family members. The expression of daDREAM in the forebrain resulted in a complex phenotype characterized by loss of recurrent inhibition and enhanced long-term potentiation (LTP) in the dentate gyrus and impaired learning and memory. Our results indicate that DREAM is a major master switch transcription factor that regulates the on/off status of specific activity-dependent gene expression programs that control synaptic plasticity, learning, and memory.


Assuntos
Regulação para Baixo/genética , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cálcio/metabolismo , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Giro Denteado/metabolismo , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Aprendizagem , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Prosencéfalo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Behav Brain Res ; 256: 291-7, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23973755

RESUMO

Carnitine palmitoyltransferase 1c (CPT1C), a brain-specific protein localized in the endoplasmic reticulum of neurons, is expressed in almost all brain regions, but its only known functions to date are involved in the hypothalamic control of energy homeostasis and in hippocampus-dependent spatial learning. To identify other physiological and behavioral functions of this protein, we performed a battery of neurological tests on Cpt1c-deficient mice. The animals showed intact autonomic and sensory systems, but some motor disturbances were observed. A more detailed study of motor function revealed impaired coordination and gait, severe muscle weakness, and reduced daily locomotor activity. Analysis of motor function in these mice at ages of 6-24 weeks showed that motor disorders were already present in young animals and that impairment increased progressively with age. Analysis of CPT1C expression in different motor brain areas during development revealed that CPT1C levels were low from birth to postnatal day 10 and then rapidly increased peaking at postnatal day 21, which suggests that CPT1C plays a relevant role in motor function during and after weaning. As CPT1C is known to regulate ceramide levels, we measured these biolipids in different motor areas in adult mice. Cerebellar, striatum, and motor cortex extracts from Cpt1c knockout mice showed reduced levels of ceramide and its derivative sphingosine when compared to wild-type animals. Our results indicate that altered ceramide metabolism in motor brain areas induced by Cpt1c deficiency causes progressive motor dysfunction from a young age.


Assuntos
Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Carnitina O-Palmitoiltransferase/deficiência , Transtornos dos Movimentos/fisiopatologia , Animais , Animais Recém-Nascidos , Western Blotting , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Ceramidas/metabolismo , Progressão da Doença , Marcha/fisiologia , Masculino , Camundongos Knockout , Atividade Motora/fisiologia , Debilidade Muscular/fisiopatologia , Exame Neurológico , Reflexo/fisiologia , Teste de Desempenho do Rota-Rod , Esfingosina/metabolismo
17.
Front Mol Neurosci ; 5: 58, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22563308

RESUMO

Downstream regulatory element antagonist modulator (DREAM) is a Ca(2+)-binding protein that binds DNA and represses transcription in a Ca(2+)-dependent manner. Previous work has shown a role for DREAM in cerebellar function regulating the expression of the sodium/calcium exchanger 3 (NCX3) in cerebellar granular neurons to control Ca(2+) homeostasis and survival of these neurons. To achieve a global view of the genes regulated by DREAM in the cerebellum, we performed a genome-wide analysis in transgenic cerebellum expressing a Ca(2+)-insensitive/CREB-independent dominant active mutant DREAM (daDREAM). Here we show that DREAM regulates the expression of the midline 1 (Mid1) gene early after birth. As a consequence, daDREAM mice exhibit a significant shortening of the rostro-caudal axis of the cerebellum and a delay in neuromotor development early after birth. Our results indicate a role for DREAM in cerebellar function.

18.
Neuropsychopharmacology ; 36(12): 2395-405, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21814183

RESUMO

Despite the impact of schizophrenia and mood disorders, which in extreme cases can lead to death, recent decades have brought little progress in the development of new treatments. Recent studies have shown that Reelin, an extracellular protein that is critical for neuronal development, is reduced in schizophrenia and bipolar disorder patients. However, data on a causal or protective role of Reelin in psychiatric diseases is scarce. In order to study the direct influence of Reelin's levels on behavior, we subjected two mouse lines, in which Reelin levels are either reduced (Reelin heterozygous mice) or increased (Reelin overexpressing mice), to a battery of behavioral tests: open-field, black-white box, novelty-suppressed-feeding, forced-swim-test, chronic corticosterone treatment followed by forced-swim-test, cocaine sensitization and pre-pulse inhibition (PPI) deficits induced by N-methyl-D-aspartate (NMDA) antagonists. These tests were designed to model some aspects of psychiatric disorders such as schizophrenia, mood, and anxiety disorders. We found no differences between Reeler heterozygous mice and their wild-type littermates. However, Reelin overexpression in the mouse forebrain reduced the time spent floating in the forced-swim-test in mice subjected to chronic corticosterone treatment, reduced behavioral sensitization to cocaine, and reduced PPI deficits induced by a NMDA antagonist. In addition, we demonstrate that while stress increased NMDA NR2B-mediated synaptic transmission, known to be implicated in depression, Reelin overexpression significantly reduced it. Together, these results point to the Reelin signaling pathway as a relevant drug target for the treatment of a range of psychiatric disorders.


Assuntos
Transtorno Bipolar/prevenção & controle , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/biossíntese , Fenótipo , Esquizofrenia/prevenção & controle , Serina Endopeptidases/biossíntese , Animais , Transtorno Bipolar/genética , Transtorno Bipolar/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Cocaína/administração & dosagem , Corticosterona/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Proteínas da Matriz Extracelular/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteína Reelina , Esquizofrenia/genética , Esquizofrenia/metabolismo , Filtro Sensorial/efeitos dos fármacos , Filtro Sensorial/fisiologia , Serina Endopeptidases/genética
19.
Neurobiol Dis ; 24(2): 403-18, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16963267

RESUMO

Accumulating evidence has suggested that neurotrophins participate in the pathophysiology of mood disorders. We have developed transgenic mice overexpressing the full-length neurotrophin-3 receptor TrkC (TgNTRK3) in the central nervous system. TgNTRK3 mice show increased anxiety-like behavior and enhancement of panic reaction in the mouse defense test battery, along with an increase in the number and density of catecholaminergic (tyrosine hydroxylase positive) neurons in locus coeruleus and substantia nigra. Furthermore, treatment of TgNTRK3 mice with diazepam significantly attenuated the anxiety-like behaviors in the plus maze. These results provide evidence for the involvement of TrkC in the development of noradrenergic neurons in the central nervous system with consequences on anxiety-like behavior and panic reaction. Thus, changes in TrkC expression levels could contribute to the phenotypic expression of panic disorder through a trophic effect on noradrenergic neurons in the locus coeruleus. Our results demonstrate that the elevated NT3-TrkC tone via overexpression of TrkC in the brain may constitute a molecular mechanism for the expression of anxiety and anxiety.


Assuntos
Transtornos de Ansiedade/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Catecolaminas/metabolismo , Neurônios/metabolismo , Transtorno de Pânico/metabolismo , Receptor trkC/metabolismo , Animais , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/fisiopatologia , Doenças do Sistema Nervoso Autônomo/genética , Doenças do Sistema Nervoso Autônomo/metabolismo , Doenças do Sistema Nervoso Autônomo/fisiopatologia , Comportamento Animal/fisiologia , Encéfalo/patologia , Contagem de Células , Proliferação de Células , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença/genética , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Locus Cerúleo/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Testes Neuropsicológicos , Norepinefrina/metabolismo , Transtorno de Pânico/genética , Transtorno de Pânico/fisiopatologia , Receptor trkC/genética , Substância Negra/metabolismo , Substância Negra/patologia , Substância Negra/fisiopatologia , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa