Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 9(5): e91613, 2014.
Article in English | MEDLINE | ID: mdl-24838000

ABSTRACT

Mutations in cyclin-dependent kinase-like 5 (CDKL5) cause early-onset epileptic encephalopathy, a neurodevelopmental disorder with similarities to Rett Syndrome. Here we describe the physiological, molecular, and behavioral phenotyping of a Cdkl5 conditional knockout mouse model of CDKL5 disorder. Behavioral analysis of constitutive Cdkl5 knockout mice revealed key features of the human disorder, including limb clasping, hypoactivity, and abnormal eye tracking. Anatomical, physiological, and molecular analysis of the knockout uncovered potential pathological substrates of the disorder, including reduced dendritic arborization of cortical neurons, abnormal electroencephalograph (EEG) responses to convulsant treatment, decreased visual evoked responses (VEPs), and alterations in the Akt/rpS6 signaling pathway. Selective knockout of Cdkl5 in excitatory and inhibitory forebrain neurons allowed us to map the behavioral features of the disorder to separable cell-types. These findings identify physiological and molecular deficits in specific forebrain neuron populations as possible pathological substrates in CDKL5 disorder.


Subject(s)
Disease Models, Animal , Phenotype , Prosencephalon/pathology , Protein Serine-Threonine Kinases/genetics , Rett Syndrome/genetics , Rett Syndrome/pathology , Signal Transduction/physiology , Spasms, Infantile/genetics , Spasms, Infantile/pathology , Analysis of Variance , Animals , Blotting, Western , Dendrites/pathology , Electroencephalography , Epileptic Syndromes , Evoked Potentials, Visual/physiology , Eye Movements/physiology , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Statistics, Nonparametric
2.
Mediators Inflamm ; 2014: 560120, 2014.
Article in English | MEDLINE | ID: mdl-24757286

ABSTRACT

Rett syndrome (RTT) is a pervasive neurodevelopmental disorder mainly linked to mutations in the gene encoding the methyl-CpG-binding protein 2 (MeCP2). Respiratory dysfunction, historically credited to brainstem immaturity, represents a major challenge in RTT. Our aim was to characterize the relationships between pulmonary gas exchange abnormality (GEA), upper airway obstruction, and redox status in patients with typical RTT (n = 228) and to examine lung histology in a Mecp2-null mouse model of the disease. GEA was detectable in ~80% (184/228) of patients versus ~18% of healthy controls, with "high" (39.8%) and "low" (34.8%) patterns dominating over "mixed" (19.6%) and "simple mismatch" (5.9%) types. Increased plasma levels of non-protein-bound iron (NPBI), F2-isoprostanes (F2-IsoPs), intraerythrocyte NPBI (IE-NPBI), and reduced and oxidized glutathione (i.e., GSH and GSSG) were evidenced in RTT with consequently decreased GSH/GSSG ratios. Apnea frequency/severity was positively correlated with IE-NPBI, F2-IsoPs, and GSSG and negatively with GSH/GSSG ratio. A diffuse inflammatory infiltrate of the terminal bronchioles and alveoli was evidenced in half of the examined Mecp2-mutant mice, well fitting with the radiological findings previously observed in RTT patients. Our findings indicate that GEA is a key feature of RTT and that terminal bronchioles are a likely major target of the disease.


Subject(s)
Inflammation/pathology , Lung Diseases/physiopathology , Mutation , Rett Syndrome/physiopathology , Adolescent , Adult , Animals , Antioxidants/metabolism , Child , Child, Preschool , Disease Models, Animal , Female , Glutathione/metabolism , Humans , Infant , Lung/pathology , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Pulmonary Gas Exchange , Rett Syndrome/metabolism , Young Adult
3.
Hum Mol Genet ; 20(6): 1182-96, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21212100

ABSTRACT

Rett syndrome (RTT) is a neurodevelopmental disorder with no efficient treatment that is caused in the majority of cases by mutations in the gene methyl-CpG binding-protein 2 (MECP2). RTT becomes manifest after a period of apparently normal development and causes growth deceleration, severe psychomotor impairment and mental retardation. Effective animal models for RTT are available and show morphofunctional abnormalities of synaptic connectivity. However, the molecular consequences of MeCP2 disruption leading to neuronal and synaptic alterations are not known. Protein synthesis regulation via the mammalian target of the rapamycin (mTOR) pathway is crucial for synaptic organization, and its disruption is involved in a number of neurodevelopmental diseases. We investigated the phosphorylation of the ribosomal protein (rp) S6, whose activation is highly dependent from mTOR activity. Immunohistochemistry showed that rpS6 phosphorylation is severely affected in neurons across the cortical areas of Mecp2 mutants and that this alteration precedes the severe symptomatic phase of the disease. Moreover, we found a severe defect of the initiation of protein synthesis in the brain of presymptomatic Mecp2 mutant that was not restricted to a specific subset of transcripts. Finally, we provide evidence for a general dysfunction of the Akt/mTOR, but not extracellular-regulated kinase, signaling associated with the disease progression in mutant brains. Our results indicate that defects in the AKT/mTOR pathway are responsible for the altered translational control in Mecp2 mutant neurons and disclosed a novel putative biomarker of the pathological process. Importantly, this study provides a novel context of therapeutic interventions that can be designed to successfully restrain or ameliorate the development of RTT.


Subject(s)
Down-Regulation , Oncogene Protein v-akt/metabolism , Protein Biosynthesis , Rett Syndrome/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Humans , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oncogene Protein v-akt/genetics , Rett Syndrome/genetics , TOR Serine-Threonine Kinases/genetics
4.
Biol Psychiatry ; 67(7): 657-65, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20172507

ABSTRACT

BACKGROUND: Rett syndrome (RTT) is an X-linked progressive neurodevelopmental disorder characterized by a variety of symptoms including motor abnormalities, mental retardation, anxiety, and autism. Most of RTT cases are caused by mutations of MeCP2. In mice, impaired MeCP2 function results in synaptic deficits associated with motor, cognitive, and emotional alterations. Environmental enrichment (EE) is a rearing condition that enhances synapse formation and plasticity. Previous studies analyzing the effects of postweaning EE found limited effects on motor performance of male MeCP2 mutants. However, EE during early postnatal development produces powerful effects on neural development and plasticity. Thus, we tested whether early EE could ameliorate several phenotypes of male homozygous and female heterozygous MeCP2 mutants. METHODS: We investigated the effects of early EE on motor coordination, structural and functional synaptic plasticity, and brain-derived neurotrophic factor expression in male MeCP2 null mice. Anxiety-related behavior and spatial learning was analyzed in heterozygous MeCP2 female mice. RESULTS: In male mutants, EE modified excitatory and to a lesser extent inhibitory synaptic density in cerebellum and cortex, reversed the cortical long-term potentiation deficit and augmented cortical brain-derived neurotrophic factor levels. Environmental enrichment also ameliorated motor coordination and motor learning. In female heterozygous mice, a model closely mimicking some aspects of RTT symptoms, EE rescued memory deficits in the Morris water maze and decreased anxiety-related behavior. CONCLUSIONS: Early EE dramatically improves several phenotypes of MeCP2 mutants. Thus, environmental factors should be taken into account when analyzing phenotypes of MeCP2 knockout mice, an accepted model of RTT. Early EE might be beneficial in RTT patients.


Subject(s)
Behavior, Animal , Environment , Methyl-CpG-Binding Protein 2/genetics , Phenotype , Synapses , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Disease Models, Animal , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Rett Syndrome/genetics , Rett Syndrome/metabolism
5.
Article in English | MEDLINE | ID: mdl-21423514

ABSTRACT

There is mounting evidence showing that the structural and molecular organization of synaptic connections is affected both in human patients and in animal models of neurological and psychiatric diseases. As a consequence of these experimental observations, it has been introduced the concept of synapsopathies, a notion describing brain disorders of synaptic function and plasticity. A close correlation between neurological diseases and synaptic abnormalities is especially relevant for those syndromes including also mental retardation in their symptomatology, such as Rett syndrome (RS). RS (MIM312750) is an X-linked dominant neurological disorder that is caused in the majority of cases by mutations in methyl-CpG-binding protein 2 (MeCP2). This review will focus on the current knowledge of the synaptic alterations produced by mutations of the gene MeCP2 in mouse models of RS and will highlight prospects experimental therapies currently in use. Different experimental approaches have revealed that RS could be the consequence of an impairment in the homeostasis of synaptic transmission in specific brain regions. Indeed, several forms of experience-induced neuronal plasticity are impaired in the absence of MeCP2. Based on the results presented in this review, it is reasonable to propose that understanding how the brain is affected by diseases such as RS is at reach. This effort will bring us closer to identify the neurobiological bases of human cognition.

6.
Neuron ; 53(5): 747-59, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17329213

ABSTRACT

The action of visual experience on visual cortical circuits is maximal during a critical period of postnatal development. The long-term effects of this experience are likely mediated by signaling cascades regulating experience-dependent gene transcription. Developmental modifications of these pathways could explain the difference in plasticity between the young and adult cortex. We studied the pathways linking experience-dependent activation of ERK to CREB-mediated gene expression in vivo. In juvenile mice, visual stimulation that activates CREB-mediated gene transcription also induced ERK-dependent MSK and histone H3 phosphorylation and H3-H4 acetylation, an epigenetic mechanism of gene transcription activation. In adult animals, ERK and MSK were still inducible; however, visual stimulation induced weak CREB-mediated gene expression and H3-H4 posttranslational modifications. Stimulation of histone acetylation in adult animals by means of trichostatin promoted ocular dominance plasticity. Thus, differing, experience-dependent activations of signaling molecules might be at the basis of the differences in experience-dependent plasticity between juvenile and adult cortex.


Subject(s)
Histones/metabolism , Neuronal Plasticity/physiology , Protein Processing, Post-Translational/physiology , Visual Cortex/physiology , Acetylation/drug effects , Age Factors , Animals , Critical Period, Psychological , Cyclic AMP Response Element-Binding Protein/metabolism , Dominance, Cerebral/physiology , Down-Regulation/genetics , Epigenesis, Genetic/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Histones/genetics , Hydroxamic Acids/pharmacology , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Phosphorylation , Photic Stimulation , Protein Synthesis Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Visual Cortex/cytology , Visual Cortex/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...