Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cereb Cortex ; 34(1)2024 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-37997361

RESUMO

In this study, we investigated the impact of Dual specificity tyrosine-phosphorylation-regulated kinase 1A (Dyrk1A) overexpression, a gene associated with Down syndrome, on hippocampal neuronal deficits in mice. Our findings revealed that mice overexpressing Dyrk1A (TgDyrk1A; TG) exhibited impaired hippocampal recognition memory, disrupted excitation-inhibition balance, and deficits in long-term potentiation (LTP). Specifically, we observed layer-specific deficits in dendritic arborization of TG CA1 pyramidal neurons in the stratum radiatum. Through computational modeling, we determined that these alterations resulted in reduced storage capacity and compromised integration of inputs, with decreased high γ oscillations. Contrary to prevailing assumptions, our model suggests that deficits in neuronal architecture, rather than over-inhibition, primarily contribute to the reduced network. We explored the potential of environmental enrichment (EE) as a therapeutic intervention and found that it normalized the excitation-inhibition balance, restored LTP, and improved short-term recognition memory. Interestingly, we observed transient significant dendritic remodeling, leading to recovered high γ. However, these effects were not sustained after EE discontinuation. Based on our findings, we conclude that Dyrk1A overexpression-induced layer-specific neuromorphological disturbances impair the encoding of place and temporal context. These findings contribute to our understanding of the underlying mechanisms of Dyrk1A-related hippocampal deficits and highlight the challenges associated with long-term therapeutic interventions for cognitive impairments.


Assuntos
Proteínas Serina-Treonina Quinases , Proteínas Tirosina Quinases , Animais , Camundongos , Modelos Animais de Doenças , Camundongos Transgênicos , Neurônios , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Células Piramidais
2.
Cell Mol Life Sci ; 80(12): 370, 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37989807

RESUMO

Individuals with Down syndrome (DS) have a higher prevalence of obesity compared to the general population. Conventionally, this has been attributed to endocrine issues and lack of exercise. However, deficits in neural reward responses and dopaminergic disturbances in DS may be contributing factors. To investigate this, we focused on a mouse model (Ts65Dn) bearing some triplicated genes homologous to trisomy 21. Through detailed meal pattern analysis in male Ts65Dn mice, we observed an increased preference for energy-dense food, pointing towards a potential "hedonic" overeating behavior. Moreover, trisomic mice exhibited higher scores in compulsivity and inflexibility tests when limited access to energy-dense food and quinine hydrochloride adulteration were introduced, compared to euploid controls. Interestingly, when we activated prelimbic-to-nucleus accumbens projections in Ts65Dn male mice using a chemogenetic approach, impulsive and compulsive behaviors significantly decreased, shedding light on a promising intervention avenue. Our findings uncover a novel mechanism behind the vulnerability to overeating and offer potential new pathways for tackling obesity through innovative interventions.


Assuntos
Síndrome de Down , Trissomia , Humanos , Masculino , Camundongos , Animais , Síndrome de Down/genética , Modelos Animais de Doenças , Córtex Pré-Frontal , Hiperfagia/genética , Obesidade/genética
3.
bioRxiv ; 2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-36945607

RESUMO

Abnormal dendritic arbors, dendritic spine "dysgenesis" and excitation inhibition imbalance are main traits assumed to underlie impaired cognition and behavioral adaptation in intellectual disability. However, how these modifications actually contribute to functional properties of neuronal networks, such as signal integration or storage capacity is unknown. Here, we used a mouse model overexpressing Dyrk1A (Dual-specificity tyrosine [Y]-regulated kinase), one of the most relevant Down syndrome (DS) candidate genes, to gather quantitative data regarding hippocampal neuronal deficits produced by the overexpression of Dyrk1A in mice (TgDyrk1A; TG). TG mice showed impaired hippocampal recognition memory, altered excitation-inhibition balance and deficits in hippocampal CA1 LTP. We also detected for the first time that deficits in dendritic arborization in TG CA1 pyramidal neurons are layer-specific, with a reduction in the width of the stratum radiatum, the postsynaptic target site of CA3 excitatory neurons, but not in the stratum lacunosum-moleculare, which receives temporo-ammonic projections. To interrogate about the functional impact of layer-specific TG dendritic deficits we developed tailored computational multicompartmental models. Computational modelling revealed that neuronal microarchitecture alterations in TG mice lead to deficits in storage capacity, altered the integration of inputs from entorhinal cortex and hippocampal CA3 region onto CA1 pyramidal cells, important for coding place and temporal context and on connectivity and activity dynamics, with impaired the ability to reach high γ oscillations. Contrary to what is assumed in the field, the reduced network activity in TG is mainly contributed by the deficits in neuronal architecture and to a lesser extent by over-inhibition. Finally, given that therapies aimed at improving cognition have also been tested for their capability to recover dendritic spine deficits and excitation-inhibition imbalance, we also tested the short- and long-term changes produced by exposure to environmental enrichment (EE). Exposure to EE normalized the excitation inhibition imbalance and LTP, and had beneficial effects on short-term recognition memory. Importantly, it produced massive but transient dendritic remodeling of hippocampal CA1, that led to recovery of high γ oscillations, the main readout of synchronization of CA1 neurons, in our simulations. However, those effects where not stable and were lost after EE discontinuation. We conclude that layer-specific neuromorphological disturbances produced by Dyrk1A overexpression impair coding place and temporal context. Our results also suggest that treatments targeting structural plasticity, such as EE, even though hold promise towards improved treatment of intellectual disabilities, only produce temporary recovery, due to transient dendritic remodeling.

4.
Mol Neurobiol ; 57(7): 3195-3205, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32504418

RESUMO

Alcoholism is a chronic relapsing disorder defined by loss of control over excessive consumption of ethanol despite damaging effects on the liver and brain. We previously showed that the overexpression in mice of Dyrk1A (TgDyrk1A, for dual-specificity tyrosine (Y) phosphorylation-regulated kinase 1A) reduces the severity of alcohol mediated liver injury. Ethanol consumption has also been associated with increased brain glutamate concentration that led to therapies targeting glutamatergic receptors and normalization of glutamatergic neurotransmission. Interestingly, mice overexpressing Dyrk1A (TgDyrk1A mice) present a reduction of glutamatergic brain transmission, which we propose could be protective against alcohol intake. To answer this question, we investigated the ethanol preference in TgDyrk1A mice using a two-bottle choice paradigm. TgDyrk1A mice showed a non-significant decrease of voluntary ethanol intake and ethanol preference compared with wild-type mice. At the peripheral level, mice overexpressing Dyrk1A show lower ethanol plasma levels, indicating a faster ethanol metabolism. At the end of the protocol, lasting 21 days, brains were extracted for protein analysis. Ethanol reduced levels of the synaptic protein PSD-95 and increased the glutamate decarboxylase GAD65, specifically in the cortex of TgDyrk1A mice. Our results suggest that overexpression of DYRK1A may cause different ethanol-induced changes in the brain.


Assuntos
Encéfalo/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Etanol/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Sinapses/metabolismo , Animais , Encéfalo/metabolismo , Comportamento de Escolha/efeitos dos fármacos , Proteína 4 Homóloga a Disks-Large/metabolismo , Glutamato Descarboxilase/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Quinases Dyrk
5.
Neuropsychopharmacology ; 42(2): 462-472, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27534266

RESUMO

The inability to properly extinguish fear memories constitutes the foundation of several anxiety disorders, including panic disorder. Recent findings show that boosting prefrontal cortex synaptic plasticity potentiates fear extinction, suggesting that therapies that augment synaptic plasticity could prove useful in rescue of fear extinction impairments in this group of disorders. Previously, we reported that mice with selective deregulation of neurotrophic tyrosine kinase receptor, type 3 expression (TgNTRK3) exhibit increased fear memories accompanied by impaired extinction, congruent with an altered activation pattern of the amygdala-hippocampus-medial prefrontal cortex fear circuit. Here we explore the specific role of neurotrophin 3 and its cognate receptor in the medial prefrontal cortex, and its involvement in fear extinction in a pathological context. In this study we combined molecular, behavioral, in vivo pharmacology and ex vivo electrophysiological recordings in TgNTRK3 animals during contextual fear extinction processes. We show that neurotrophin 3 protein levels are increased upon contextual fear extinction in wild-type animals but not in TgNTRK3 mice, which present deficits in infralimbic long-term potentiation. Importantly, infusion of neurotrophin 3 to the medial prefrontal cortex of TgNTRK3 mice rescues contextual fear extinction and ex vivo local application improves medial prefrontal cortex synaptic plasticity. This effect is blocked by inhibition of extracellular signal-regulated kinase phosphorylation through peripheral administration of SL327, suggesting that rescue occurs via this pathway. Our results suggest that stimulating neurotrophin 3-dependent medial prefrontal cortex plasticity could restore contextual fear extinction deficit in pathological fear and could constitute an effective treatment for fear-related disorders.


Assuntos
Extinção Psicológica/efeitos dos fármacos , Medo , Neurotrofina 3/administração & dosagem , Transtornos Fóbicos/fisiopatologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptor trkC/agonistas , Animais , Modelos Animais de Doenças , Extinção Psicológica/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Potenciação de Longa Duração , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasticidade Neuronal , Neurotrofina 3/fisiologia , Transtornos Fóbicos/prevenção & controle , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Receptor trkC/genética , Receptor trkC/fisiologia
6.
Mol Nutr Food Res ; 58(2): 278-88, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24039182

RESUMO

SCOPE: Trisomy for human chromosome 21 results in Down syndrome (DS), which is among the most complex genetic perturbations leading to intellectual disability. Accumulating data suggest that overexpression of the dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1A (DYRK1A), is a critical pathogenic mechanisms in the intellectual deficit. METHODS AND RESULTS: Here we show that the green tea flavonol epigallocatechin-gallate (EGCG), a DYRK1A inhibitor, rescues the cognitive deficits of both segmental trisomy 16 (Ts65Dn) and transgenic mice overexpressing Dyrk1A in a trisomic or disomic genetic background, respectively. It also significantly reverses cognitive deficits in a pilot study in DS individuals with effects on memory recognition, working memory and quality of life. We used the mouse models to ensure that EGCG was able to reduce DYRK1A kinase activity in the hippocampus and found that it also induced significant changes in plasma homocysteine levels, which were correlated with Dyrk1A expression levels. Thus, we could use plasma homocysteine levels as an efficacy biomarker in our human study. CONCLUSION: We conclude that EGCG is a promising therapeutic tool for cognitive enhancement in DS, and its efficacy may depend of Dyrk1A inhibition.


Assuntos
Catequina/análogos & derivados , Cognição/efeitos dos fármacos , Síndrome de Down/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Adolescente , Adulto , Animais , Biomarcadores/sangue , Catequina/administração & dosagem , Cromossomos Humanos Par 16/genética , Modelos Animais de Doenças , Método Duplo-Cego , Feminino , Regulação da Expressão Gênica , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mosaicismo , Fosforilação , Projetos Piloto , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Trissomia/genética , Adulto Jovem , Quinases Dyrk
7.
Amino Acids ; 45(1): 113-31, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23604404

RESUMO

The development of strategies capable to promote nervous system plasticity in adulthood is nowadays an important aim in neuroscience to improve not only cognitive abilities but also to ameliorate pathological dysfunctions. Several studies have demonstrated that adult neurogenesis is regulated by many physiological and pathological stimuli at almost every stage, from proliferation of neuronal precursors until integration and activation of newly formed neurons in the preexisting network. We review the process of generating functional neurons from precursors in the adult brain and its implications in intellectual disability disorders.


Assuntos
Encéfalo/fisiologia , Hipocampo/fisiologia , Deficiência Intelectual/fisiopatologia , Neurogênese , Animais , Diferenciação Celular , Humanos , Camundongos , Neurônios/fisiologia
8.
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.

9.
CNS Neurol Disord Drug Targets ; 9(4): 429-38, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20522013

RESUMO

Individuals with trisomy 21, also known as Down syndrome (DS), develop a clinical syndrome including almost identical neuropathological characteristics of Alzheimer's disease (AD) observed in non-DS individuals. The main difference is the early age of onset of AD pathology in individuals with DS, with hish incidence of clinical symptoms in the late 40- early 50 years of age. The neuropathology of AD in persons with DS is superimposed with the developmental abnormalities causing alterations of neuronal morphology and function. Despite the ubiquitous occurrence of AD neuropathology, clinical signs of dementia do not occur in all adults with DS even at older ages. Phenotype analysis of DS mouse models has revealed a differential age-related neurodegenerative pattern that correlates with specific biochemical and molecular alterations at the cellular level. In fact, several individual genes found in trisomy in DS have been functionally related to neuronal degeneration. Thus, mouse models overexpressing HSA21 gene(s) are fundamental to understand the neurodegenerative process in DS, as described in the present review. In addition, these models might allow to define and evaluate potential drug targets and to develop therapeutic strategies that may interfere or delay the onset of AD.


Assuntos
Modelos Animais de Doenças , Síndrome de Down/patologia , Doenças Neurodegenerativas/patologia , Adulto , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Síndrome de Down/genética , Genes/genética , Humanos , Camundongos , Camundongos Transgênicos/crescimento & desenvolvimento , Pessoa de Meia-Idade , Doenças Neurodegenerativas/genética
10.
Neurobiol Dis ; 36(2): 312-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19660545

RESUMO

Dual-specificity tyrosine-regulated kinase 1A (DYRK1A) is a candidate gene for the Down syndrome neurological defects and may be involved in the progression of Alzheimer's disease. Heterozygous mice for Dyrk1A (Dyrk1A+/-) exhibit decreased brain size, motor abnormalities and cognitive deficits in the adult. However, there is no information about the mutant phenotype in old ages. Here we analyze the impact of Dyrk1A dosage reduction on motor performance and hippocampal-dependent learning and memory in aged Dyrk1A+/- mice. Whereas motor tests showed marked alterations in traction ability, prehensile reflex and balance, heterozygous mice only present a slight impairment of visuo-spatial memory even though they show a robust decrease of CA1-CA3 and dentate gyrus cells.


Assuntos
Envelhecimento/genética , Triagem de Portadores Genéticos , Aprendizagem/fisiologia , Atividade Motora/genética , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Animais , Encéfalo/patologia , Transtornos Cognitivos/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes Neurológicos , Mutação/fisiologia , Tamanho do Órgão/genética , Desempenho Psicomotor/fisiologia , Comportamento Espacial/fisiologia , Quinases Dyrk
11.
J Cell Biol ; 178(6): 1039-51, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17785520

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal and incurable muscle degenerative disorder. We identify a function of the protease urokinase plasminogen activator (uPA) in mdx mice, a mouse model of DMD. The expression of uPA is induced in mdx dystrophic muscle, and the genetic loss of uPA in mdx mice exacerbated muscle dystrophy and reduced muscular function. Bone marrow (BM) transplantation experiments revealed a critical function for BM-derived uPA in mdx muscle repair via three mechanisms: (1) by promoting the infiltration of BM-derived inflammatory cells; (2) by preventing the excessive deposition of fibrin; and (3) by promoting myoblast migration. Interestingly, genetic loss of the uPA receptor in mdx mice did not exacerbate muscular dystrophy in mdx mice, suggesting that uPA exerts its effects independently of its receptor. These findings underscore the importance of uPA in muscular dystrophy.


Assuntos
Distrofia Muscular de Duchenne/metabolismo , Mioblastos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/deficiência , Animais , Transplante de Medula Óssea , Movimento Celular , Células Cultivadas , Fibrina/metabolismo , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Mioblastos/patologia , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase
12.
Behav Genet ; 36(3): 454-68, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16520905

RESUMO

Trisomy 21 or Down syndrome (DS) is a complex syndrome, of genetic origin with multiple and variable neurobiological and neuropsychological manifestations. DS patients have consistent signs of brain damage along their lives, but understanding the biology of DS is complicated due to the extraordinary heterogeneity of the phenotypic signs. Thus, treatment of DS mental retardation poses significant challenges for clinicians and scientists. The review addresses the classical pharmacological and environmental treatments and also critically reviews the new possibilities that are emerging from the exciting advances in gene or cell therapy. We describe some of the most recent developments in the field and give a sense of the prospects for future prevention and therapy.


Assuntos
Síndrome de Down/terapia , Medicina Baseada em Evidências/métodos , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Encéfalo/patologia , Modelos Animais de Doenças , Síndrome de Down/tratamento farmacológico , Síndrome de Down/genética , Medicina Baseada em Evidências/tendências , Humanos , Lactente , Camundongos , N-Metilaspartato/fisiologia , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...