Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 289(15): 10831-10842, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24567331

ABSTRACT

The ability of a neuron to transduce extracellular signals into long lasting changes in neuronal morphology is central to its normal function. Increasing evidence shows that coordinated regulation of synaptic and nuclear signaling in response to NMDA receptor activation is crucial for long term memory, synaptic tagging, and epigenetic signaling. Although mechanisms have been proposed for synapse-to-nuclear communication, it is unclear how signaling is coordinated at both subcompartments. Here, we show that activation of NMDA receptors induces the bi-directional and concomitant shuttling of the scaffold protein afadin from the cytosol to the nucleus and synapses. Activity-dependent afadin nuclear translocation peaked 2 h post-stimulation, was independent of protein synthesis, and occurred concurrently with dendritic spine remodeling. Moreover, activity-dependent afadin nuclear translocation coincides with phosphorylation of histone H3 at serine 10 (H3S10p), a marker of epigenetic modification. Critically, blocking afadin nuclear accumulation attenuated activity-dependent dendritic spine remodeling and H3 phosphorylation. Collectively, these data support a novel model of neuronal nuclear signaling whereby dual-residency proteins undergo activity-dependent bi-directional shuttling from the cytosol to synapses and the nucleus, coordinately regulating dendritic spine remodeling and histone modifications.


Subject(s)
Cell Nucleus/metabolism , Dendritic Spines/metabolism , Histones/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Synapses/metabolism , Active Transport, Cell Nucleus , Animals , Brain/embryology , Cytosol/metabolism , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Neuronal Plasticity/physiology , Neurons/metabolism , Phosphorylation , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Signal Transduction
2.
J Biol Chem ; 287(43): 35964-74, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22948147

ABSTRACT

The dendritic field of a neuron, which is determined by both dendritic architecture and synaptic strength, defines the synaptic input of a cell. Once established, a neuron's dendritic field is thought to remain relatively stable throughout a cell's lifetime. Perturbations in a dendritic structure or excitatory tone of a cell and thus its dendritic field are cellular alterations thought to be correlated with a number of psychiatric disorders. Although several proteins are known to regulate the development of dendritic arborization, much less is known about the mechanisms that maintain dendritic morphology and synaptic strength. In this study, we find that afadin, a component of N-cadherin·ß-catenin·α-N-catenin adhesion complexes, is required for the maintenance of established dendritic arborization and synapse number. We further demonstrate that afadin directly interacts with AMPA receptors and that loss of this protein reduces the surface expression of GluA1- and GluA2-AMPA receptor subunits. Collectively, these data suggest that afadin is required for the maintenance of dendritic structure and excitatory tone.


Subject(s)
Dendrites/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Dendrites/genetics , Gene Expression Regulation/physiology , LIM Domain Proteins/genetics , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Synapses/genetics , alpha Catenin/genetics , alpha Catenin/metabolism , beta Catenin/genetics , beta Catenin/metabolism
3.
J Neurochem ; 123(5): 781-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22862288

ABSTRACT

Dendritic spines serve as the post-synaptic structural component of synapses. The structure and function of dendritic spines are dynamically regulated by a number of signaling pathways and allow for normal neural processing, whereas aberrant spine changes are thought to contribute to cognitive impairment in neuropsychiatric and neurodegenerative disorders. However, spine changes within different brain regions and their contribution to specific cognitive functions, especially later in adulthood, is not well understood. In this study, we used late-adult KALRN-deficient mice as a tool to investigate the vulnerability of different cognitive functions to long-term perturbations in spine plasticity in different forebrain regions. We found that in these mice, loss of one or both copies of KALRN lead to genotype and brain region-dependent reductions in spine density. Surprisingly, heterozygote and knockout mice showed differential impairments in cognitive phenotypes, including working memory, social recognition, and social approach. Correlation analysis between the site and magnitude of spine loss and behavioral alterations suggests that the interplay between brain regions is critical for complex cognitive processing and underscores the importance of spine plasticity in normal cognitive function. Long-term perturbation of spine plasticity results in distinct impairments of cognitive function. Using genetically modified mice deficient in a central regulator of spine plasticity, we investigated the brain region-specific contribution of spine numbers to various cognitive functions. We found distinct cognitive functions display differential sensitivity to spine loss in the cortex and hippocampus. Our data support spines as neuronal structures important for cognition and suggest interplay between brain regions is critical for complex cognitive processing.


Subject(s)
Brain/pathology , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Dendritic Spines/pathology , Neuronal Plasticity/physiology , Animals , Brain/physiopathology , Cognition/physiology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Male , Mice , Mice, Knockout
4.
Neurobiol Dis ; 45(2): 796-803, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22120753

ABSTRACT

Reductions in dendritic arbor length and complexity are among the most consistently replicated changes in neuronal structure in post mortem studies of cerebral cortical samples from subjects with schizophrenia, however, the underlying molecular mechanisms have not been identified. This study is the first to identify an alteration in a regulatory protein which is known to promote both dendritic length and arborization in developing neurons, Kalirin-9. We found Kalirin-9 expression to be paradoxically increased in schizophrenia. We followed up this observation by overexpressing Kalirin-9 in mature primary neuronal cultures, causing reduced dendritic length and complexity. Kalirin-9 overexpression represents a potential mechanism for dendritic changes seen in schizophrenia.


Subject(s)
Dendrites/pathology , Guanine Nucleotide Exchange Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Schizophrenia/metabolism , Schizophrenia/pathology , Adult , Animals , Auditory Cortex/metabolism , Auditory Cortex/pathology , Blotting, Western , Dendrites/metabolism , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Rats , Rats, Sprague-Dawley
5.
Brain Res Rev ; 67(1-2): 184-92, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21276817

ABSTRACT

Representing the most common cause of dementia, Alzheimer's disease (AD) has dramatically impacted the neurological and economic health of our society. AD is a debilitating neurodegenerative disease that produces marked cognitive decline. Much evidence has accumulated over the past decade to suggest soluble oligomers of beta-amyloid (Aß) have a critical role in mediating AD pathology early in the disease process by perturbing synaptic efficacy. Here we critically review recent research that implicates synapses as key sites of early pathogenesis in AD. Most excitatory synapses in the brain rely on dendritic spines as the sites for excitatory neurotransmission. The structure and function of dendritic spines are dynamically regulated by cellular pathways acting on the actin cytoskeleton. Numerous studies analyzing human postmortem tissue, animal models and cellular paradigms indicate that AD pathology has a deleterious effect on the pathways governing actin cytoskeleton stability. Based on the available evidence, we propose the idea that a contributing factor to synaptic pathology in early AD is an Aß oligomer-initiated collapse of a "synaptic safety net" in spines, leading to dendritic spine degeneration and synaptic dysfunction. Spine stabilizing pathways may thus represent efficacious therapeutic targets for combating AD pathology.


Subject(s)
Actin Cytoskeleton/pathology , Actins/toxicity , Alzheimer Disease/pathology , Synapses/pathology , Actin Cytoskeleton/chemistry , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Animals , Dendritic Spines/pathology , Humans , Synapses/chemistry , Synapses/physiology , Synaptic Transmission/physiology
6.
Nat Neurosci ; 14(3): 285-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21346746

ABSTRACT

Substantial progress has been made toward understanding the genetic architecture, cellular substrates, brain circuits and endophenotypic profiles of neuropsychiatric disorders, including autism spectrum disorders (ASD), schizophrenia and Alzheimer's disease. Recent evidence implicates spiny synapses as important substrates of pathogenesis in these disorders. Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of spine pathology may provide insight into their etiologies and may reveal new drug targets. Here we discuss recent neuropathological, genetic, molecular and animal model studies that implicate structural alterations at spiny synapses in the pathogenesis of major neurological disorders, focusing on ASD, schizophrenia and Alzheimer's disease as representatives of these categories across different ages of onset. We stress the importance of reverse translation, collaborative and multidisciplinary approaches, and the study of the spatio-temporal roles of disease molecules in the context of synaptic regulatory pathways and neuronal circuits that underlie disease endophenotypes.


Subject(s)
Dendritic Spines/pathology , Nervous System Diseases/pathology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Schizophrenia/genetics , Schizophrenia/pathology , Schizophrenia/physiopathology
7.
J Neurosci Res ; 88(3): 650-68, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19746427

ABSTRACT

Dopamine depletion leads to impaired motor performance and increased glutamatergic-mediated hyperexcitability of medium spiny neurons in the basal ganglia. Intensive treadmill exercise improves motor performance in both saline treatment and the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. In the present study, we investigated the effect of high-intensity treadmill exercise on changes in alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit expression, because these receptor channels confer the majority of fast excitatory neurotransmission in the brain, and their subunit composition provides a key mechanism for regulating synaptic strength and synaptic neuroplasticity and is important in modulating glutamatergic neurotransmission. Within the dorsolateral striatum of MPTP mice, treadmill exercise increased GluR2 subunit expression, with no significant effect on GluR1. Furthermore, neurophysiological studies demonstrated a reduction in the size of excitatory postsynaptic currents (EPSCs) in striatal medium spiny neurons (as determined by the input-output relationship), reduced amplitude of spontaneous EPSCs, and a loss of polyamine-sensitive inward rectification, all supportive of an increase in heteromeric AMPAR channels containing the GluR2 subunit. Phosphorylation of GluR2 at serine 880 in both saline-treated and MPTP mice suggests that exercise may also influence AMPAR trafficking and thus synaptic strength within the striatum. Finally, treadmill exercise also altered flip isoforms of GluR2 and GluR1 mRNA transcripts. These findings suggest a role for AMPARs in mediating the beneficial effects of exercise and support the idea that adaptive changes in GluR2 subunit expression may be important in modulating experience-dependent neuroplasticity of the injured basal ganglia.


Subject(s)
Basal Ganglia/injuries , Basal Ganglia/physiopathology , Parkinson Disease, Secondary/physiopathology , Physical Conditioning, Animal/physiology , Receptors, AMPA/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Excitatory Postsynaptic Potentials/physiology , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Neurons/physiology , Parkinson Disease, Secondary/chemically induced , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Random Allocation , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...