Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
eNeuro ; 4(6)2017.
Article in English | MEDLINE | ID: mdl-29333487

ABSTRACT

Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by loss-of-function mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2; Amir et al., 1999), a transcriptional regulatory protein (Klose et al., 2005). Mouse models of RTT (Mecp2 mutants) exhibit excitatory hypoconnectivity in the medial prefrontal cortex (mPFC; Sceniak et al., 2015), a region critical for functions that are abnormal in RTT patients, ranging from learning and memory to regulation of visceral homeostasis (Riga et al., 2014). The present study was designed to test the hypothesis that increasing the activity of mPFC pyramidal neurons in heterozygous female Mecp2 mutants (Hets) would ameliorate RTT-like symptoms, including deficits in respiratory control and long-term retrieval of auditory conditioned fear. Selective activation of mPFC pyramidal neurons in adult animals was achieved by bilateral infection with an AAV8 vector expressing excitatory hm3D(Gq) DREADD (Designer Receptors Exclusively Activated by Designer Drugs) (Armbruster et al., 2007) under the control of the CamKIIa promoter. DREADD activation in Mecp2 Hets completely restored long-term retrieval of auditory conditioned fear, eliminated respiratory apneas, and reduced respiratory frequency variability to wild-type (Wt) levels. Reversal of respiratory symptoms following mPFC activation was associated with normalization of Fos protein levels, a marker of neuronal activity, in a subset of brainstem respiratory neurons. Thus, despite reduced levels of MeCP2 and severe neurological deficits, mPFC circuits in Het mice are sufficiently intact to generate normal behavioral output when pyramidal cell activity is increased. These findings highlight the contribution of mPFC hypofunction to the pathophysiology of RTT and raise the possibility that selective activation of cortical regions such as the mPFC could provide therapeutic benefit to RTT patients.


Subject(s)
Cognition/physiology , Prefrontal Cortex/physiopathology , Pyramidal Cells/physiology , Respiration , Rett Syndrome/physiopathology , Animals , Auditory Perception/physiology , Conditioning, Psychological/physiology , Designer Drugs , Disease Models, Animal , Fear/physiology , Female , Genetic Vectors , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Transgenic , Random Allocation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
2.
J Neurosci ; 34(49): 16369-84, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471575

ABSTRACT

NG2 is purportedly one of the most growth-inhibitory chondroitin sulfate proteoglycans (CSPGs) produced after spinal cord injury. Nonetheless, once the severed axon tips dieback from the lesion core into the penumbra they closely associate with NG2+ cells. We asked if proteoglycans play a role in this tight cell-cell interaction and whether overadhesion upon these cells might participate in regeneration failure in rodents. Studies using varying ratios of CSPGs and adhesion molecules along with chondroitinase ABC, as well as purified adult cord-derived NG2 glia, demonstrate that CSPGs are involved in entrapping neurons. Once dystrophic axons become stabilized upon NG2+ cells, they form synaptic-like connections both in vitro and in vivo. In NG2 knock-out mice, sensory axons in the dorsal columns dieback further than their control counterparts. When axons are double conditioned to enhance their growth potential, some traverse the lesion core and express reduced amounts of synaptic proteins. Our studies suggest that proteoglycan-mediated entrapment upon NG2+ cells is an additional obstacle to CNS axon regeneration.


Subject(s)
Antigens/physiology , Axons/physiology , Cell Communication/physiology , Nerve Regeneration/physiology , Proteoglycans/physiology , Spinal Cord Injuries/physiopathology , Synapses/physiology , Animals , Antigens/genetics , Axons/ultrastructure , Cell Tracking , Cells, Cultured , Chondroitin Sulfate Proteoglycans/physiology , Fibronectins/physiology , Ganglia, Spinal/physiopathology , Ganglia, Spinal/ultrastructure , Integrin beta1/physiology , Laminin/physiology , Mice , Mice, Knockout , Nerve Degeneration/physiopathology , Proteoglycans/genetics
3.
Dis Model Mech ; 5(6): 733-45, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23115203

ABSTRACT

In September of 2011, the National Institute of Neurological Disorders and Stroke (NINDS), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), the International Rett Syndrome Foundation (IRSF) and the Rett Syndrome Research Trust (RSRT) convened a workshop involving a broad cross-section of basic scientists, clinicians and representatives from the National Institutes of Health (NIH), the US Food and Drug Administration (FDA), the pharmaceutical industry and private foundations to assess the state of the art in animal studies of Rett syndrome (RTT). The aim of the workshop was to identify crucial knowledge gaps and to suggest scientific priorities and best practices for the use of animal models in preclinical evaluation of potential new RTT therapeutics. This review summarizes outcomes from the workshop and extensive follow-up discussions among participants, and includes: (1) a comprehensive summary of the physiological and behavioral phenotypes of RTT mouse models to date, and areas in which further phenotypic analyses are required to enhance the utility of these models for translational studies; (2) discussion of the impact of genetic differences among mouse models, and methodological differences among laboratories, on the expression and analysis, respectively, of phenotypic traits; and (3) definitions of the standards that the community of RTT researchers can implement for rigorous preclinical study design and transparent reporting to ensure that decisions to initiate costly clinical trials are grounded in reliable preclinical data.


Subject(s)
Rett Syndrome/pathology , Translational Research, Biomedical , Animals , Congresses as Topic , Disease Models, Animal , Guidelines as Topic , Humans , Research Report , Rett Syndrome/genetics
4.
J Neurosci ; 32(40): 13860-72, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035095

ABSTRACT

Excitatory-inhibitory imbalance has been identified within specific brain microcircuits in models of Rett syndrome (RTT) and other autism spectrum disorders (ASDs). However, macrocircuit dysfunction across the RTT brain as a whole has not been defined. To approach this issue, we mapped expression of the activity-dependent, immediate-early gene product Fos in the brains of wild-type (Wt) and methyl-CpG-binding protein 2 (Mecp2)-null (Null) mice, a model of RTT, before and after the appearance of overt symptoms (3 and 6 weeks of age, respectively). At 6 weeks, Null mice exhibit significantly less Fos labeling than Wt in limbic cortices and subcortical structures, including key nodes in the default mode network. In contrast, Null mice exhibit significantly more Fos labeling than Wt in the hindbrain, most notably in cardiorespiratory regions of the nucleus tractus solitarius (nTS). Using nTS as a model, whole-cell recordings demonstrated that increased Fos expression in Nulls at 6 weeks of age is associated with synaptic hyperexcitability, including increased frequency of spontaneous and miniature EPSCs and increased amplitude of evoked EPSCs in Nulls. No such effect of genotype on Fos or synaptic function was seen at 3 weeks. In the mutant forebrain, reduced Fos expression, as well as abnormal sensorimotor function, were reversed by the NMDA receptor antagonist ketamine. In light of recent findings that the default mode network is hypoactive in autism, our data raise the possibility that hypofunction within this meta-circuit is a shared feature of RTT and other ASDs and is reversible.


Subject(s)
Autonomic Nervous System/physiopathology , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Methyl-CpG-Binding Protein 2/physiology , Nerve Net/physiopathology , Prosencephalon/physiopathology , Solitary Nucleus/physiopathology , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , Genes, fos , Humans , Male , Methyl-CpG-Binding Protein 2/deficiency , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Nerve Net/drug effects , Nerve Net/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Organ Specificity , Patch-Clamp Techniques , Prosencephalon/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Rett Syndrome/genetics , Rett Syndrome/physiopathology , Sensory Gating/drug effects , Sensory Gating/physiology , Solitary Nucleus/chemistry , Solitary Nucleus/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...