Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 9(30): eadi0286, 2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37506203

RESUMO

Polypyrimidine tract binding protein 1 (PTBP1) is thought to be expressed only at embryonic stages in central neurons. Its down-regulation triggers neuronal differentiation in precursor and non-neuronal cells, an approach recently tested for generation of neurons de novo for amelioration of neurodegenerative disorders. Moreover, PTBP1 is replaced by its paralog PTBP2 in mature central neurons. Unexpectedly, we found that both proteins are coexpressed in adult sensory and motor neurons, with PTBP2 restricted mainly to the nucleus, while PTBP1 also shows axonal localization. Levels of axonal PTBP1 increased markedly after peripheral nerve injury, and it associates in axons with mRNAs involved in injury responses and nerve regeneration, including importin ß1 (KPNB1) and RHOA. Perturbation of PTBP1 affects local translation in axons, nociceptor neuron regeneration and both thermal and mechanical sensation. Thus, PTBP1 has functional roles in adult axons. Hence, caution is required before considering targeting of PTBP1 for therapeutic purposes.


Assuntos
Axônios , Regeneração Nervosa , Neurônios , Traumatismos dos Nervos Periféricos , Adulto , Humanos , Axônios/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Interneurônios/metabolismo , Regeneração Nervosa/genética , Neurônios/metabolismo , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo
2.
Dev Med Child Neurol ; 65(2): 162-170, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056801

RESUMO

Rett syndrome (RTT) is an X-linked neurogenetic disorder caused by mutations of the MECP2 (methyl-CpG-binding protein 2) gene. Over two decades of work established MeCP2 as a protein with pivotal roles in the regulation of the epigenome, neuronal physiology, synaptic maintenance, and behaviour. Given the genetic aetiology of RTT and the proof of concept of its reversal in a mouse model, considerable efforts have been made to design therapeutic approaches to re-express MeCP2. By being at the forefront of the development of innovative gene therapies, research on RTT is of paramount importance for the treatment of monogenic neurological diseases. Here we discuss the recent advances and challenges of promising genetic strategies for the treatment of RTT including gene replacement therapies, gene/RNA editing strategies, and reactivation of the silenced X chromosome. WHAT THIS PAPER ADDS: Recent advances shed light on the promises of gene replacement therapy with new vectors designed to control the levels of MeCP2 expression. New developments in DNA/RNA editing approaches or reactivation of the silenced X chromosome open the possibility to re-express the native MeCP2 locus at endogenous levels. Current strategies still face limitations in transduction efficiency and future work is needed to improve brain delivery.


Assuntos
Arteterapia , Síndrome de Rett , Camundongos , Animais , Humanos , Síndrome de Rett/terapia , Síndrome de Rett/tratamento farmacológico , Proteína 2 de Ligação a Metil-CpG/genética , Encéfalo/metabolismo , Mutação , Neurônios
3.
Cell Rep Med ; 2(5): 100281, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34095883

RESUMO

Anxiety and stress-related conditions represent a significant health burden in modern society. Unfortunately, most anxiolytic drugs are prone to side effects, limiting their long-term usage. Here, we employ a bioinformatics screen to identify drugs for repurposing as anxiolytics. Comparison of drug-induced gene-expression profiles with the hippocampal transcriptome of an importin α5 mutant mouse model with reduced anxiety identifies the hypocholesterolemic agent ß-sitosterol as a promising candidate. ß-sitosterol activity is validated by both intraperitoneal and oral application in mice, revealing it as the only clear anxiolytic from five closely related phytosterols. ß-sitosterol injection reduces the effects of restraint stress, contextual fear memory, and c-Fos activation in the prefrontal cortex and dentate gyrus. Moreover, synergistic anxiolysis is observed when combining sub-efficacious doses of ß-sitosterol with the SSRI fluoxetine. These preclinical findings support further development of ß-sitosterol, either as a standalone anxiolytic or in combination with low-dose SSRIs.


Assuntos
Ansiolíticos/farmacologia , Transtornos de Ansiedade/tratamento farmacológico , Ansiedade/tratamento farmacológico , Sitosteroides/farmacologia , Animais , Medo/efeitos dos fármacos , Fluoxetina/farmacologia , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Preparações Farmacêuticas/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Tranquilizantes/farmacologia
4.
Science ; 369(6505): 842-846, 2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32792398

RESUMO

How is neuropathic pain regulated in peripheral sensory neurons? Importins are key regulators of nucleocytoplasmic transport. In this study, we found that importin α3 (also known as karyopherin subunit alpha 4) can control pain responsiveness in peripheral sensory neurons in mice. Importin α3 knockout or sensory neuron-specific knockdown in mice reduced responsiveness to diverse noxious stimuli and increased tolerance to neuropathic pain. Importin α3-bound c-Fos and importin α3-deficient neurons were impaired in c-Fos nuclear import. Knockdown or dominant-negative inhibition of c-Fos or c-Jun in sensory neurons reduced neuropathic pain. In silico screens identified drugs that mimic importin α3 deficiency. These drugs attenuated neuropathic pain and reduced c-Fos nuclear localization. Thus, perturbing c-Fos nuclear import by importin α3 in peripheral neurons can promote analgesia.


Assuntos
Dor Crônica/fisiopatologia , Neuralgia/fisiopatologia , Células Receptoras Sensoriais/fisiologia , alfa Carioferinas/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Benzofenonas/farmacologia , Dor Crônica/genética , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Isoxazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/genética , Proteínas Proto-Oncogênicas c-fos/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fator de Transcrição AP-1/metabolismo , alfa Carioferinas/genética
5.
EMBO Mol Med ; 12(2): e10889, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31913581

RESUMO

Mutations in the X-linked MECP2 gene are responsible for Rett syndrome (RTT), a severe neurological disorder for which there is no treatment. Several studies have linked the loss of MeCP2 function to alterations of brain-derived neurotrophic factor (BDNF) levels, but non-specific overexpression of BDNF only partially improves the phenotype of Mecp2-deficient mice. We and others have previously shown that huntingtin (HTT) scaffolds molecular motor complexes, transports BDNF-containing vesicles, and is under-expressed in Mecp2 knockout brains. Here, we demonstrate that promoting HTT phosphorylation at Ser421, either by a phospho-mimetic mutation or inhibition of the phosphatase calcineurin, restores endogenous BDNF axonal transport in vitro in the corticostriatal pathway, increases striatal BDNF availability and synaptic connectivity in vivo, and improves the phenotype and the survival of Mecp2 knockout mice-even though treatments were initiated only after the mice had already developed symptoms. Stimulation of endogenous cellular pathways may thus be a promising approach for the treatment of RTT patients.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Proteína Huntingtina/química , Proteína 2 de Ligação a Metil-CpG , Síndrome de Rett/genética , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Feminino , Homeostase , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fosforilação
6.
Neuron ; 102(3): 507-509, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31071280

RESUMO

In this issue of Neuron, Crerar et al. (2019) found Tp53inp2 as a highly expressed RNA in sympathetic neuron axons. Strikingly, its long 3' UTR ensures that Tp53inp2 is not translated in axons, and the untranslated RNA affects neuronal growth by interacting with neurotrophin receptors.


Assuntos
Fator de Crescimento Neural , RNA , Axônios , RNA Mensageiro , Transdução de Sinais
7.
Cell Rep ; 25(11): 3169-3179.e7, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30540948

RESUMO

Importins mediate transport from synapse to soma and from cytoplasm to nucleus, suggesting that perturbation of importin-dependent pathways should have significant neuronal consequences. A behavioral screen on five importin α knockout lines revealed that reduced expression of importin α5 (KPNA1) in hippocampal neurons specifically decreases anxiety in mice. Re-expression of importin α5 in ventral hippocampus of knockout animals increased anxiety behaviors to wild-type levels. Hippocampal neurons lacking importin α5 reveal changes in presynaptic plasticity and modified expression of MeCP2-regulated genes, including sphingosine kinase 1 (Sphk1). Knockout of importin α5, but not importin α3 or α4, reduces MeCP2 nuclear localization in hippocampal neurons. A Sphk1 blocker reverses anxiolysis in the importin α5 knockout mouse, while pharmacological activation of sphingosine signaling has robust anxiolytic effects in wild-type animals. Thus, importin α5 influences sphingosine-sensitive anxiety pathways by regulating MeCP2 nuclear import in hippocampal neurons.


Assuntos
Ansiedade/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , alfa Carioferinas/metabolismo , Animais , Ansiolíticos/farmacologia , Comportamento Animal , Carbolinas/farmacologia , Hipocampo/patologia , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Sinapses/metabolismo , Transcrição Gênica , alfa Carioferinas/deficiência
8.
J Neurosci Methods ; 270: 9-16, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27269190

RESUMO

BACKGROUND: Behavioral analyses in rodents have successfully delineated the function of many genes and signaling pathways in the brain. Behavioral testing uses highly defined experimental conditions to identify abnormalities in a given mouse strain or genotype. The open field (OF) is widely used to assess both locomotion and anxiety in rodents. In this test, the more a mouse explores and spend time in the center of the arena, the less anxious it is considered to be. However, the simplistic distinction between center and border substantially reduces the information content of the analysis and may fail to detect biologically meaningful differences. NEW METHOD: Here we describe COLORcation, a new application for improved analyses of mouse behavior in the OF. RESULTS: The application analyses animal exploration patterns in detailed spatial resolution (e.g. 10×10 bins) to provide a color-encoded heat map of mouse activity. In addition, COLORcation provides new parameters to track activity and locomotion of the test animals. We demonstrate the use of COLORcation in different experimental paradigms, including pharmacological and restraint-based induction of stress and anxiety. COMPARISON WITH EXISTING METHOD(S): COLORcation is compatible with multiple acquisition systems, giving users the option to make the most of their raw data organized text files containing time and coordinates of animal locations as input. CONCLUSION: These analyses validate the utility of the software and establish its reliability and potential as a new tool to analyze OF data.


Assuntos
Algoritmos , Ansiedade , Comportamento Exploratório , Análise de Variância , Animais , Ansiedade/fisiopatologia , Carbolinas , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora , Fenótipo , Restrição Física , Software , Estresse Psicológico
9.
Trends Neurosci ; 38(2): 108-16, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25534890

RESUMO

Local signaling events at synapses or axon terminals must be communicated to the nucleus to elicit transcriptional responses. The lengths of neuronal processes pose a significant challenge for such intracellular communication. This challenge is met by mechanisms ranging from rapid signals encoded in calcium waves to slower macromolecular signaling complexes carried by molecular motors. Here we summarize recent findings on macromolecular signaling from the synapse to the nucleus, in comparison to those employed in injury signaling along axons. A number of common themes emerge, including combinatorial signal encoding by post-translational mechanisms such as differential phosphorylation and proteolysis, and conserved roles for importins in coordinating signaling complexes. Neurons may integrate ionic flux with motor-transported signals as a temporal code for synaptic plasticity signaling.


Assuntos
Transporte Biológico/fisiologia , Núcleo Celular/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Animais , Axônios/fisiologia , Humanos
10.
PLoS One ; 9(3): e92169, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24667344

RESUMO

Proper brain functioning requires a fine-tuning between excitatory and inhibitory neurotransmission, a balance maintained through the regulation and release of glutamate and GABA. Rett syndrome (RTT) is a rare genetic disorder caused by mutations in the methyl-CpG binding protein 2 (MECP2) gene affecting the postnatal brain development. Dysfunctions in the GABAergic and glutamatergic systems have been implicated in the neuropathology of RTT and a disruption of the balance between excitation and inhibition, together with a perturbation of the electrophysiological properties of GABA and glutamate neurons, were reported in the brain of the Mecp2-deficient mouse. However, to date, the extent and the nature of the GABA/glutamate deficit affecting the Mecp2-deficient mouse brain are unclear. In order to better characterize these deficits, we simultaneously analyzed the GABA and glutamate levels in Mecp2-deficient mice at 2 different ages (P35 and P55) and in several brain areas. We used a multilevel approach including the quantification of GABA and glutamate levels, as well as the quantification of the mRNA and protein expression levels of key genes involved in the GABAergic and glutamatergic pathways. Our results show that Mecp2-deficient mice displayed regional- and age-dependent variations in the GABA pathway and, to a lesser extent, in the glutamate pathway. The implication of the GABA pathway in the RTT neuropathology was further confirmed using an in vivo treatment with a GABA reuptake inhibitor that significantly improved the lifespan of Mecp2-deficient mice. Our results confirm that RTT mouse present a deficit in the GABAergic pathway and suggest that GABAergic modulators could be interesting therapeutic agents for this severe neurological disorder.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Ácido Glutâmico/metabolismo , Proteína 2 de Ligação a Metil-CpG/fisiologia , Síndrome de Rett/metabolismo , Transdução de Sinais , Ácido gama-Aminobutírico/metabolismo , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Agonistas GABAérgicos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácidos Nipecóticos/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Síndrome de Rett/etiologia , Síndrome de Rett/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transmissão Sináptica , Tiagabina , Ácido gama-Aminobutírico/química , Ácido gama-Aminobutírico/genética
11.
Brain ; 136(Pt 10): 3018-27, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24030951

RESUMO

The central nervous system has a pattern of gene expression that is closely regulated with respect to functional and anatomical regions. DNA methylation is a major regulator of transcriptional activity, and aberrations in the distribution of this epigenetic mark may be involved in many neurological disorders, such as Alzheimer's disease. Herein, we have analysed 12 distinct mouse brain regions according to their CpG 5'-end gene methylation patterns and observed their unique epigenetic landscapes. The DNA methylomes obtained from the cerebral cortex were used to identify aberrant DNA methylation changes that occurred in two mouse models of Alzheimer's disease. We were able to translate these findings to patients with Alzheimer's disease, identifying DNA methylation-associated silencing of three targets genes: thromboxane A2 receptor (TBXA2R), sorbin and SH3 domain containing 3 (SORBS3) and spectrin beta 4 (SPTBN4). These hypermethylation targets indicate that the cyclic AMP response element-binding protein (CREB) activation pathway and the axon initial segment could contribute to the disease.


Assuntos
Doença de Alzheimer/genética , Encéfalo/metabolismo , Metilação de DNA/genética , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , DNA/genética , Epigênese Genética/genética , Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/genética
12.
F1000Res ; 2: 204, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24555100

RESUMO

Rett syndrome is a neurological disorder caused by mutations in the MECP2 gene.  MeCP2 transcripts are alternatively spliced to generate two protein isoforms (MeCP2_e1 and MeCP2_e2) that differ at their N-termini. Whilst mRNAs for both forms are expressed ubiquitously, the one for MeCP2_e1 is more abundant than for MeCP2_e2 in the central nervous system. In transfected cells, both protein isoforms are nuclear and colocalize with densely methylated heterochromatic foci. With a view to understanding the physiological contribution of each isoform, and their respective roles in the pathogenesis of Rett syndrome, we set out to generate isoform-specific anti-MeCP2 antibodies. To this end, we immunized rabbits against the peptides corresponding to the short amino-terminal portions that are different between the two isoforms. The polyclonal antibodies thus obtained specifically detected their respective isoforms of MeCP2 in Neuro2a (N2A) cells transfected to express either form. Both antisera showed comparable sensitivities when used for Western blot or immunofluorescence, and were highly specific for their respective isoform. When those antibodies were used on mouse tissues, specific signals were easily detected for Mecp2_e1, whilst Mecp2_e2 was very difficult to detect by Western blot, and even more so by immunofluorescence. Our results thus suggest that brain cells express low amounts of the Mecp2-e2 isoform. Our findings are compatible with recent reports showing that MeCP2_e2 is dispensable for healthy brain function, and that it may be involved in the regulation of neuronal apoptosis and embryonic development.

14.
Neurobiol Dis ; 45(2): 786-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22127389

RESUMO

Mecp2 deficiency or overexpression causes a wide spectrum of neurological diseases in humans among which Rett Syndrome is the prototype. Pathogenic mechanisms are thought to involve transcriptional deregulation of target genes such as Bdnf together with defects in the general transcriptional program of affected cells. Here we found that two master genes, Huntingtin (Htt) and huntingtin-associated protein (Hap1), involved in the control of Bdnf axonal transport, are altered in the brain of Mecp2-deficient mice. We also revealed an in vivo defect of Bdnf transport throughout the cortico striatal pathway of Mecp2-deficient animals. We found that the velocity of Bdnf-containing vesicles is reduced in vitro in the Mecp2-deficient axons and this deficit can be rescued by the re-expression of Mecp2. The defect in axonal transport is not restricted to Bdnf since transport of the amyloid precursor protein (App) that is Htt and Hap1-dependent is also altered. Finally, treating Mecp2-deficient mice with cysteamine, a molecule increasing the secretion of Bdnf vesicles, improved the lifespan and reduced motor defects, suggesting a new therapeutic strategy for Rett syndrome.


Assuntos
Transporte Axonal/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Imunofluorescência , Perfilação da Expressão Gênica , Proteína Huntingtina , Imuno-Histoquímica , Masculino , Proteína 2 de Ligação a Metil-CpG/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Transfecção
15.
BMC Neurosci ; 12: 47, 2011 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-21609470

RESUMO

BACKGROUND: Rett syndrome (RTT, MIM #312750) is a severe neurological disorder caused by mutations in the X-linked methyl-CpG binding protein 2 (MECP2) gene. Female patients are affected with an incidence of 1/15000 live births and develop normally from birth to 6-18 months of age before the onset of deficits in autonomic, cognitive, motor functions (stereotypic hand movements, impaired locomotion) and autistic features. Studies on Mecp2 mouse models, and specifically null mice, revealed morphological and functional alterations of neurons. Several functions that are regulated by bioaminergic nuclei or peripheral ganglia are impaired in the absence of Mecp2. RESULTS: Using high performance liquid chromatography, combined with electrochemical detection (HPLC/EC) we found that Mecp2(-/y) mice exhibit an alteration of DA metabolism in the ponto-bulbar region at 5 weeks followed by a more global alteration of monoamines when the disease progresses (8 weeks). Hypothalamic measurements suggest biphasic disturbances of norepinephrine and serotonin at pathology onset (5 weeks) that were found stabilized later on (8 weeks). Interestingly, the postnatal nigrostriatal dopaminergic deficit identified previously does not parallel the reduction of the other neurotransmitters investigated. Finally, dosage in cortical samples do not suggest modification in the monoaminergic content respectively at 5 and 8 weeks of age. CONCLUSIONS: We have identified that the level of catecholamines and serotonin is differentially affected in Mecp2(-/y) brain areas in a time-dependent fashion.


Assuntos
Encéfalo/metabolismo , Dopamina/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Norepinefrina/metabolismo , Serotonina/metabolismo , Fatores Etários , Animais , Cromatografia Líquida de Alta Pressão , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Knockout
16.
Behav Brain Res ; 216(1): 313-20, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20713094

RESUMO

The methyl-CpG binding protein 2 (Mecp2) gene encodes a nuclear transcriptional modulator highly expressed in post-mitotic neurons. Mutations of this gene cause a large spectrum of neurological disorders in humans. Several lines of mice harboring a constitutional deletion of Mecp2 are available. The use of these models is crucial to understand the basis of Mecp2-related pathologies. However, most of the studies performed using these lines focused on different postnatal time points. The aim of the present study was to provide a more complete description of the behavioral phenotype of the Mecp2(tm1.1Bird) mice. To this aim, we used a modified version of the SHIRPA protocol and a set of sensorimotor tests and respiratory metabolism measurements, in a longitudinal study of the Mecp2-null male mice (Mecp2(-/y)) from three weeks (weaning) to eight weeks of age. Our data document, for the first time, the sequential appearance of the in vivo deficits in this mouse line. The observed deficits initially concern major parameters (such as body weight), and are followed by involuntary and sensitive defects (reflexes). Subsequently, motor functions and respiratory metabolism are severally impaired. A detailed description of these gradual defects may help to identify their neuronal origin and to elaborate novel therapeutic strategies.


Assuntos
Proteína 2 de Ligação a Metil-CpG/genética , Atividade Motora/genética , Fadiga Muscular/genética , Consumo de Oxigênio/genética , Respiração/genética , Análise de Variância , Animais , Força da Mão/fisiologia , Masculino , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fenótipo , Teste de Desempenho do Rota-Rod , Estatísticas não Paramétricas
17.
Neurobiol Dis ; 41(2): 385-97, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20951208

RESUMO

Rett syndrome (RTT) is a severe neurological disorder caused by mutations in the MECP2 gene, in which older patients often develop parkinsonian features. Although Mecp2 has been shown to modulate the catecholaminergic metabolism of the RTT mouse model, little is known about the central dopaminergic neurons. Here we found that the progression of the motor dysfunction in the Mecp2-deficient mouse becomes more severe between 4 and 9 weeks of age. We then studied the phenotype of the dopaminergic neurons of the substantia nigra pars compacta (SNpc). We found a major reduction in the number of tyrosine hydroxylase (Th)-expressing neurons, as well as a reduction in their soma size, by 5 weeks of age. We showed that this deficit is not due to apoptosis and that the remaining neurons express a mature dopaminergic phenotype. A reduction in the Th-staining intensity was also found in the caudate-putamen (CPu), the main dopaminergic target for SNpc. We found that the amount of activated-Th (pSer40-Th) is slightly reduced at 5 weeks of age in the Mecp2-deficient mouse, but that this amount is affected more importantly by 9 weeks of age. Neurochemical measurements revealed a significant reduction of dopamine content at 5 and 9 weeks of age in the CPu whereas SNpc contents were preserved. Finally, we found that chronic L-Dopa treatment improved the motor deficits previously identified. Altogether, our findings demonstrate that Mecp2-deficiency induces nigrostriatal deficits, and they offer a new perspective to better understand the origin of motor dysfunction in RTT.


Assuntos
Proteína 2 de Ligação a Metil-CpG/deficiência , Proteína 2 de Ligação a Metil-CpG/genética , Síndrome de Rett/patologia , Síndrome de Rett/fisiopatologia , Substância Negra/patologia , Substância Negra/fisiopatologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Dopamina/deficiência , Masculino , Proteína 2 de Ligação a Metil-CpG/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/patologia , Neurônios Motores/fisiologia , Fenótipo , Síndrome de Rett/genética , Substância Negra/metabolismo
18.
Proc Natl Acad Sci U S A ; 107(3): 1196-200, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20080543

RESUMO

Deep brain stimulation (DBS) is a reversible technique that is currently used for the treatment of Parkinson disease and may be suitable for the treatment of psychiatric disorders. Whether DBS inactivates the target structure is still a matter of debate. Here, from findings obtained in rats, we propose DBS of the subthalamic nucleus (STN) as a possible treatment for cocaine addiction to be further tested in human studies. We show that STN DBS reversibly reduces the motivation to work for an i.v. injection of cocaine, and it increases motivation to work for sucrose pellets. These opposite effects may result from STN DBS effect on the positive affective properties of these rewards. Indeed, we further show that STN DBS reduces the preference for a place previously associated with the rewarding properties of cocaine, and it increases the preference for a place associated with food. Because these findings are consistent with those observed after STN lesions [Baunez C, Dias C, Cador M, Amalric M (2005) Nat Neurosci 8:484-489], they suggest that STN DBS mimics an inactivation of the STN on motivational processes. Furthermore, given that one of the major challenges for cocaine addiction is to find a treatment that reduces the craving for the drug without diminishing the motivation for naturally rewarding activities, our findings validate STN as a good target and DBS as the appropriate technique for a promising therapeutic strategy in the treatment of cocaine addiction.


Assuntos
Cocaína/administração & dosagem , Estimulação Encefálica Profunda , Motivação , Núcleo Subtalâmico/fisiologia , Análise de Variância , Animais , Masculino , Ratos , Ratos Long-Evans
19.
J Neurosci Res ; 88(7): 1500-9, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19998492

RESUMO

Methyl-CpG binding protein 2 (MeCP2) is a transcriptional regulator. Mutations in this gene cause a wide range of neurological disorders. Mecp2 deficiency has been previously associated to catecholaminergic dysfunctions leading to autonomic defects in the brainstem and the sympathoadrenergic system of the mouse. The present study was undertaken to determine if the locus coeruleus (LC), the main noradrenergic cell group of the brain, is affected. Using real type PCR, we found a reduction of the tyrosine hydroxylase (Th) mRNA level, the rate-limiting enzyme in catecholamine synthesis, in the whole pons of P15 (-36%), P30 (-47%) and P50 (-42%) Mecp2 null male as well as in adult heterozygous female (-44%) mice. Using immunoquantification we did not observe any difference of the Th staining level in P30 null male mice. However at P50, we demonstrated a significant decrease in both the Th staining level (-24%), and the number of Th-positive neurons (-23%). We subsequently characterized a reduction (-28%) of the dendritic density of the Th-positive fibers surrounding the LC in P50 null male mice. In heterozygous female mice immunoquantification did not revealed significant modifications, but only a tendency towards reduction. Finally, we did not found any apoptotic neurons in the pons indicating that LC neurons are not dying but are more likely loosing their catecholaminergic phenotype. In conclusion, our results showing a progressive catecholaminergic deficit in the LC of Mecp2 deficient null male mice could open new perspectives to better understand the autonomic and cognitive deficits due to the lack of Mecp2.


Assuntos
Locus Cerúleo/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Norepinefrina/deficiência , Animais , Axônios/metabolismo , Axônios/patologia , Contagem de Células , Dendritos/metabolismo , Dendritos/patologia , Regulação para Baixo/fisiologia , Feminino , Imuno-Histoquímica , Locus Cerúleo/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Norepinefrina/biossíntese , Fenótipo , RNA Mensageiro/metabolismo , Caracteres Sexuais , Tirosina 3-Mono-Oxigenase/análise , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...