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.
Elife ; 62017 09 13.
Article in English | MEDLINE | ID: mdl-28901289

ABSTRACT

Experience and activity refine cortical circuits through synapse elimination, but little is known about the activity patterns and downstream molecular mechanisms that mediate this process. We used optogenetics to drive individual mouse CA1 hippocampal neurons to fire in theta frequency bursts to understand how cell autonomous, postsynaptic activity leads to synapse elimination. Brief (1 hr) periods of postsynaptic bursting selectively depressed AMPA receptor (R) synaptic transmission, or silenced excitatory synapses, whereas more prolonged (24 hr) firing depressed both AMPAR and NMDAR EPSCs and eliminated spines, indicative of a synapse elimination. Both synapse silencing and elimination required de novo transcription, but only silencing required the activity-dependent transcription factors MEF2A/D. Burst firing induced MEF2A/D-dependent induction of the target gene Arc which contributed to synapse silencing and elimination. This work reveals new and distinct forms of activity and transcription-dependent synapse depression and suggests that these processes can occur independently.


Subject(s)
CA1 Region, Hippocampal/physiology , Neuronal Plasticity , Synapses/metabolism , Animals , Excitatory Postsynaptic Potentials , MEF2 Transcription Factors/metabolism , Mice , Optogenetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
2.
Neuron ; 93(1): 48-56, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27989458

ABSTRACT

Development of proper cortical circuits requires an interaction of sensory experience and genetic programs. Little is known of how experience and specific transcription factors interact to determine the development of specific neocortical circuits. Here, we demonstrate that the activity-dependent transcription factor, Myocyte enhancer factor-2C (Mef2c), differentially regulates development of local versus long-range excitatory synaptic inputs onto layer 2/3 neurons in the somatosensory neocortex in vivo. Postnatal, postsynaptic deletion of Mef2c in a sparse population of L2/3 neurons suppressed development of excitatory synaptic connections from all local input pathways tested. In the same cell population, Mef2c deletion promoted the strength of excitatory inputs originating from contralateral neocortex. Both the synapse promoting and synapse suppressing effects of Mef2c deletion required normal whisking experience. These results reveal a role of Mef2c in experience-dependent development of specific sensory neocortical circuits.


Subject(s)
Neocortex/metabolism , Pyramidal Cells/metabolism , Somatosensory Cortex/metabolism , Synapses/metabolism , Animals , Gene Knockdown Techniques , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Knockout , Neocortex/growth & development , Neurons/metabolism , RNA, Messenger/metabolism , Somatosensory Cortex/growth & development , Vibrissae
3.
Neuron ; 82(3): 645-58, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24811383

ABSTRACT

Repeated cocaine exposure causes persistent, maladaptive alterations in brain and behavior, and hope for effective therapeutics lies in understanding these processes. We describe here an essential role for fragile X mental retardation protein (FMRP), an RNA-binding protein and regulator of dendritic protein synthesis, in cocaine conditioned place preference, behavioral sensitization, and motor stereotypy. Cocaine reward deficits in FMRP-deficient mice stem from elevated mGluR5 (or GRM5) function, similar to a subset of fragile X symptoms, and do not extend to natural reward. We find that FMRP functions in the adult nucleus accumbens (NAc), a critical addiction-related brain region, to mediate behavioral sensitization but not cocaine reward. FMRP-deficient mice also exhibit several abnormalities in NAc medium spiny neurons, including reduced presynaptic function and premature changes in dendritic morphology and glutamatergic neurotransmission following repeated cocaine treatment. Together, our findings reveal FMRP as a critical mediator of cocaine-induced behavioral and synaptic plasticity.


Subject(s)
Cocaine/administration & dosage , Fragile X Mental Retardation Protein/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Synapses/drug effects , Synapses/physiology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Self Administration
4.
J Neurosci ; 31(35): 12426-36, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21880903

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and its cognate receptor, TrkB, regulate a wide range of cellular processes, including dendritic spine formation and functional synapse plasticity. However, the signaling mechanisms that link BDNF-activated TrkB to F-actin remodeling enzymes and dendritic spine morphological plasticity remain poorly understood. We report here that BDNF/TrkB signaling in neurons activates the Vav family of Rac/RhoA guanine nucleotide exchange factors through a novel TrkB-dependent mechanism. We find that Vav is required for BDNF-stimulated Rac-GTP production in cortical and hippocampal neurons. Vav is partially enriched at excitatory synapses in the postnatal hippocampus but does not appear to be required for normal dendritic spine density. Rather, we observe significant reductions in both BDNF-induced, rapid, dendritic spine head growth and in CA3-CA1 theta burst-stimulated long-term potentiation in Vav-deficient mouse hippocampal slices, suggesting that Vav-dependent regulation of dendritic spine morphological plasticity facilitates normal functional synapse plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Dendritic Spines/drug effects , Neuronal Plasticity/drug effects , Neurons/cytology , Proto-Oncogene Proteins c-vav/metabolism , Synapses/drug effects , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Electric Stimulation , Embryo, Mammalian , Gene Expression Regulation, Developmental/drug effects , Green Fluorescent Proteins , Hippocampus/cytology , Humans , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Mice , Neurons/ultrastructure , Organ Culture Techniques , Rats , Synapses/physiology , Synaptosomes/drug effects , Transfection/methods
6.
Proc Natl Acad Sci U S A ; 105(47): 18561-6, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19017804

ABSTRACT

Motor learning and neuro-adaptations to drugs of abuse rely upon neuronal signaling in the striatum. Cyclin-dependent kinase 5 (Cdk5) regulates striatal dopamine neurotransmission and behavioral responses to cocaine. Although the role for Cdk5 in neurodegeneration in the cortex and hippocampus and in hippocampal-dependent learning has been demonstrated, its dysregulation in the striatum has not been examined. Here we show that strong activation of striatal NMDA receptors produced p25, the truncated form of the Cdk5 co-activator p35. Furthermore, inducible overexpression of p25 in the striatum prevented locomotor sensitization to cocaine and attenuated motor coordination and learning. This corresponded with reduced dendritic spine density, increased neuro-inflammation, altered dopamine signaling, and shifted Cdk5 specificity with regard to physiological and aberrant substrates, but no apparent loss of striatal neurons. Thus, dysregulation of Cdk5 dramatically affects striatal-dependent brain function and may be relevant to non-neurodegenerative disorders involving dopamine neurotransmission.


Subject(s)
Cocaine/pharmacology , Corpus Striatum/enzymology , Cyclin-Dependent Kinase 5/physiology , Dendrites/drug effects , Learning , Locomotion , Animals , Behavior, Animal , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dendrites/physiology , Mice , Mice, Transgenic , Receptors, N-Methyl-D-Aspartate/metabolism
7.
Neuron ; 59(4): 621-33, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18760698

ABSTRACT

Repeated exposure to cocaine causes sensitized behavioral responses and increased dendritic spines on medium spiny neurons of the nucleus accumbens (NAc). We find that cocaine regulates myocyte enhancer factor 2 (MEF2) transcription factors to control these two processes in vivo. Cocaine suppresses striatal MEF2 activity in part through a mechanism involving cAMP, the regulator of calmodulin signaling (RCS), and calcineurin. We show that reducing MEF2 activity in the NAc in vivo is required for the cocaine-induced increases in dendritic spine density. Surprisingly, we find that increasing MEF2 activity in the NAc, which blocks the cocaine-induced increase in dendritic spine density, enhances sensitized behavioral responses to cocaine. Together, our findings implicate MEF2 as a key regulator of structural synapse plasticity and sensitized responses to cocaine and suggest that reducing MEF2 activity (and increasing spine density) in NAc may be a compensatory mechanism to limit long-lasting maladaptive behavioral responses to cocaine.


Subject(s)
Cocaine/pharmacology , Dendritic Spines/drug effects , Dopamine Uptake Inhibitors/pharmacology , Myogenic Regulatory Factors/metabolism , Neuronal Plasticity/drug effects , Nucleus Accumbens/drug effects , Adaptation, Physiological/drug effects , Animals , Behavior, Animal/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Cyclin-Dependent Kinases/drug effects , Down-Regulation , Drug Administration Schedule , Gene Expression Profiling , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Neostriatum/cytology , Neostriatum/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Synapses/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...