Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
Cereb Cortex ; 30(4): 2573-2585, 2020 04 14.
Article in English | MEDLINE | ID: mdl-31800021

ABSTRACT

It is generally accepted that formation and storage of memory relies on alterations of the structure and function of brain circuits. However, the structural data, which show learning-induced and long-lasting remodeling of synapses, are still very sparse. Here, we reconstruct 1927 dendritic spines and their postsynaptic densities (PSDs), representing a postsynaptic part of the glutamatergic synapse, in the hippocampal area CA1 of the mice that underwent spatial training. We observe that in young adult (5 months), mice volume of PSDs, but not the volume of the spines, is increased 26 h after the training. The training-induced growth of PSDs is specific for the dendritic spines that lack smooth endoplasmic reticulum and spine apparatuses, and requires autophosphorylation of αCaMKII. Interestingly, aging alters training-induced ultrastructural remodeling of dendritic spines. In old mice, both the median volumes of dendritic spines and PSDs shift after training toward bigger values. Overall, our data support the hypothesis that formation of memory leaves long-lasting footprint on the ultrastructure of brain circuits; however, the form of circuit remodeling changes with age.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/enzymology , Memory, Long-Term/physiology , Post-Synaptic Density/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Dendritic Spines/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/physiology , Post-Synaptic Density/genetics , Post-Synaptic Density/ultrastructure
2.
J Biol Chem ; 293(7): 2232-2246, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29269412

ABSTRACT

Altering the expression of Tomosyn-1 (Tomo-1), a soluble, R-SNARE domain-containing protein, significantly affects behavior in mice, Drosophila, and Caenorhabditis elegans Yet, the mechanisms that modulate Tomo-1 expression and its regulatory activity remain poorly defined. Here, we found that Tomo-1 expression levels influence postsynaptic spine density. Tomo-1 overexpression increased dendritic spine density, whereas Tomo-1 knockdown (KD) decreased spine density. These findings identified a novel action of Tomo-1 on dendritic spines, which is unique because it occurs independently of Tomo-1's C-terminal R-SNARE domain. We also demonstrated that the ubiquitin-proteasome system (UPS), which is known to influence synaptic strength, dynamically regulates Tomo-1 protein levels. Immunoprecipitated and affinity-purified Tomo-1 from cultured rat hippocampal neurons was ubiquitinated, and the levels of ubiquitinated Tomo-1 dramatically increased upon pharmacological proteasome blockade. Moreover, Tomo-1 ubiquitination appeared to be mediated through an interaction with the E3 ubiquitin ligase HRD1, as immunoprecipitation of Tomo-1 from neurons co-precipitated HRD1, and this interaction increases upon proteasome inhibition. Further, in vitro reactions indicated direct, HRD1 concentration-dependent Tomo-1 ubiquitination. We also noted that the UPS regulates both Tomo-1 expression and functional output, as HRD1 KD in hippocampal neurons increased Tomo-1 protein level and dendritic spine density. Notably, the effect of HRD1 KD on spine density was mitigated by additional KD of Tomo-1, indicating a direct HRD1/Tomo-1 effector relationship. In summary, our results indicate that the UPS is likely to participate in tuning synaptic efficacy and spine dynamics by precise regulation of neuronal Tomo-1 levels.


Subject(s)
Dendritic Spines/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , R-SNARE Proteins/metabolism , Ubiquitin/metabolism , Animals , Cells, Cultured , Dendritic Spines/enzymology , Dendritic Spines/genetics , Female , Hippocampus/cytology , Hippocampus/enzymology , Male , Nerve Tissue Proteins/genetics , Neurons/enzymology , Post-Synaptic Density/genetics , Post-Synaptic Density/metabolism , Proteasome Endopeptidase Complex/genetics , Protein Binding , R-SNARE Proteins/genetics , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
Proc Natl Acad Sci U S A ; 113(26): E3619-28, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27233938

ABSTRACT

Fragile X syndrome (FXS) is caused by the absence of the Fragile X Mental Retardation Protein (FMRP) in neurons. In the mouse, the lack of FMRP is associated with an excessive translation of hundreds of neuronal proteins, notably including postsynaptic proteins. This local protein synthesis deregulation is proposed to underlie the observed defects of glutamatergic synapse maturation and function and to affect preferentially the hundreds of mRNA species that were reported to bind to FMRP. How FMRP impacts synaptic protein translation and which mRNAs are most important for the pathology remain unclear. Here we show by cross-linking immunoprecipitation in cortical neurons that FMRP is mostly associated with one unique mRNA: diacylglycerol kinase kappa (Dgkκ), a master regulator that controls the switch between diacylglycerol and phosphatidic acid signaling pathways. The absence of FMRP in neurons abolishes group 1 metabotropic glutamate receptor-dependent DGK activity combined with a loss of Dgkκ expression. The reduction of Dgkκ in neurons is sufficient to cause dendritic spine abnormalities, synaptic plasticity alterations, and behavior disorders similar to those observed in the FXS mouse model. Overexpression of Dgkκ in neurons is able to rescue the dendritic spine defects of the Fragile X Mental Retardation 1 gene KO neurons. Together, these data suggest that Dgkκ deregulation contributes to FXS pathology and support a model where FMRP, by controlling the translation of Dgkκ, indirectly controls synaptic proteins translation and membrane properties by impacting lipid signaling in dendritic spine.


Subject(s)
Diacylglycerol Kinase/metabolism , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Neurons/enzymology , Aged , Animals , Dendritic Spines/enzymology , Dendritic Spines/metabolism , Diacylglycerol Kinase/genetics , Diglycerides/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/enzymology , Fragile X Syndrome/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurons/metabolism , Signal Transduction
4.
PLoS Biol ; 13(10): e1002282, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26506154

ABSTRACT

Exposure to extreme stress can trigger the development of major depressive disorder (MDD) as well as post-traumatic stress disorder (PTSD). The molecular mechanisms underlying the structural and functional alterations within corticolimbic brain regions, including the prefrontal cortex (PFC) and amygdala of individuals subjected to traumatic stress, remain unknown. In this study, we show that serum and glucocorticoid regulated kinase 1 (SGK1) expression is down-regulated in the postmortem PFC of PTSD subjects. Furthermore, we demonstrate that inhibition of SGK1 in the rat medial PFC results in helplessness- and anhedonic-like behaviors in rodent models. These behavioral changes are accompanied by abnormal dendritic spine morphology and synaptic dysfunction. Together, the results are consistent with the possibility that altered SGK1 signaling contributes to the behavioral and morphological phenotypes associated with traumatic stress pathophysiology.


Subject(s)
Depressive Disorder, Major/etiology , Enzyme Repression , Immediate-Early Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Prefrontal Cortex/metabolism , Protein Serine-Threonine Kinases/metabolism , Stress Disorders, Post-Traumatic/metabolism , Adult , Animals , Behavior, Animal , Cohort Studies , Dendritic Spines/enzymology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Female , Gene Transfer Techniques , Hippocampus/enzymology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Male , Middle Aged , Nerve Tissue Proteins/genetics , Neurons/enzymology , Neurons/pathology , Prefrontal Cortex/enzymology , Prefrontal Cortex/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Rats, Sprague-Dawley , Signal Transduction , Stress Disorders, Post-Traumatic/pathology , Stress Disorders, Post-Traumatic/psychology , Synaptic Transmission , Tissue Banks
5.
Nature ; 472(7341): 100-4, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21423166

ABSTRACT

The Rho family of GTPases have important roles in the morphogenesis of the dendritic spines of neurons in the brain and synaptic plasticity by modulating the organization of the actin cytoskeleton. Here we used two-photon fluorescence lifetime imaging microscopy to monitor the activity of two Rho GTPases-RhoA and Cdc42-in single dendritic spines undergoing structural plasticity associated with long-term potentiation in CA1 pyramidal neurons in cultured slices of rat hippocampus. When long-term volume increase was induced in a single spine using two-photon glutamate uncaging, RhoA and Cdc42 were rapidly activated in the stimulated spine. These activities decayed over about five minutes, and were then followed by a phase of persistent activation lasting more than half an hour. Although active RhoA and Cdc42 were similarly mobile, their activity patterns were different. RhoA activation diffused out of the stimulated spine and spread over about 5 µm along the dendrite. In contrast, Cdc42 activation was restricted to the stimulated spine, and exhibited a steep gradient at the spine necks. Inhibition of the Rho-Rock pathway preferentially inhibited the initial spine growth, whereas the inhibition of the Cdc42-Pak pathway blocked the maintenance of sustained structural plasticity. RhoA and Cdc42 activation depended on Ca(2+)/calmodulin-dependent kinase (CaMKII). Thus, RhoA and Cdc42 relay transient CaMKII activation to synapse-specific, long-term signalling required for spine structural plasticity.


Subject(s)
Dendritic Spines/enzymology , Dendritic Spines/physiology , Neuronal Plasticity/physiology , rho GTP-Binding Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Enzyme Activation , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/metabolism , Humans , Learning/physiology , Long-Term Potentiation/physiology , Microscopy, Fluorescence , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Pyramidal Cells/physiology , Rats , Signal Transduction , Time Factors , rho GTP-Binding Proteins/antagonists & inhibitors , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 111(17): 6461-6, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24706880

ABSTRACT

Drug discovery in psychiatry has been limited to chemical modifications of compounds originally discovered serendipitously. Therefore, more mechanism-oriented strategies of drug discovery for mental disorders are awaited. Schizophrenia is a devastating mental disorder with synaptic disconnectivity involved in its pathophysiology. Reduction in the dendritic spine density is a major alteration that has been reproducibly reported in the cerebral cortex of patients with schizophrenia. Disrupted-in-Schizophrenia-1 (DISC1), a factor that influences endophenotypes underlying schizophrenia and several other neuropsychiatric disorders, has a regulatory role in the postsynaptic density in association with the NMDA-type glutamate receptor, Kalirin-7, and Rac1. Prolonged knockdown of DISC1 leads to synaptic deterioration, reminiscent of the synaptic pathology of schizophrenia. Thus, we tested the effects of novel inhibitors to p21-activated kinases (PAKs), major targets of Rac1, on synaptic deterioration elicited by knockdown expression of DISC1. These compounds not only significantly ameliorated the synaptic deterioration triggered by DISC1 knockdown but also partially reversed the size of deteriorated synapses in culture. One of these PAK inhibitors prevented progressive synaptic deterioration in adolescence as shown by in vivo two-photon imaging and ameliorated a behavioral deficit in prepulse inhibition in adulthood in a DISC1 knockdown mouse model. The efficacy of PAK inhibitors may have implications in drug discovery for schizophrenia and related neuropsychiatric disorders in general.


Subject(s)
Aging/pathology , Dendritic Spines/pathology , Protein Kinase Inhibitors/therapeutic use , Schizophrenia/drug therapy , Schizophrenia/enzymology , p21-Activated Kinases/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Dendritic Spines/drug effects , Dendritic Spines/enzymology , Disease Models, Animal , Gene Knockdown Techniques , Mice , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Prefrontal Cortex/physiopathology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridones/chemistry , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Interference/drug effects , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/physiopathology , Synapses/drug effects , Synapses/metabolism , p21-Activated Kinases/metabolism
7.
J Neurochem ; 139(6): 959-972, 2016 12.
Article in English | MEDLINE | ID: mdl-27207106

ABSTRACT

Since the discovery of long-term potentiation (LTP) about a half-century ago, Ca2+ /CaM-dependent protein kinase II (CaMKII) has been one of the most extensively studied components of the molecular machinery that regulate plasticity. This unique dodecameric kinase complex plays pivotal roles in LTP by phosphorylating substrates through elaborate regulatory mechanisms, and is known to be both necessary and sufficient for LTP. In addition to acting as a kinase, CaMKII has been postulated to have structural roles because of its extraordinary abundance and diverse interacting partners. It now is becoming clear that these two functions of CaMKII cooperate closely for the induction of both functional and structural synaptic plasticity of dendritic spines. Because of its extraordinary abundance within neuronal cells, calmodulin kinase CaMKII has been believed to act as a structural protein as well as an enzyme during synaptic plasticity. In this review, we summarized studies in CaMKII field and provide an insight into how enzymatic and structural functions of CaMKII cooperate with each other for long-term potentiation (LTP) in neurons. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/enzymology , Long-Term Potentiation/physiology , Animals , Dendritic Spines/enzymology , Humans , Microtubules/enzymology , Neuronal Plasticity/physiology
8.
Hippocampus ; 26(4): 502-16, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26443682

ABSTRACT

Anxiety disorders are presumably associated with negative memory. Psychological therapies are widely used to treat this mental deficit in human beings based on the view that positive memory competes with negative memory and relieves anxiety status. Cellular and molecular processes underlying psychological therapies remain elusive. Therefore, we have investigated its mechanisms based on a mouse model in which food reward at one open-arm of the elevated plus-maze was used for training mice to form reward memory and challenge the open arms. Mice with the reward training showed increased entries and stay time in reward open-arm versus neutral open-arm as well as in open-arms versus closed-arms. Accompanying with reward memory formation and anxiety relief, glutamatergic synaptic transmission in dentate gyrus in vivo and dendritic spines in granule cells became upregulated. This synaptic up-regulation was accompanied by the expression of more protein kinase C (PKC) in the dendritic spines. The inhibition of PKC by chelerythrine impaired the formation of reward memory, the relief of anxiety-related behavior and the up-regulation of glutamate synapses. Our results suggest that reward-induced positive memory relieves mouse anxiety-related behavior by strengthening synaptic efficacy and PKC in the hippocampus, which imply the underlying cellular and molecular processes involved in the beneficial effects of psychological therapies treating anxiety disorders.


Subject(s)
Anxiety Disorders/therapy , Dentate Gyrus/enzymology , Memory/physiology , Protein Kinase C/metabolism , Reward , Synapses/enzymology , Animals , Anxiety Disorders/enzymology , Anxiety Disorders/pathology , Anxiety Disorders/psychology , Benzophenanthridines/pharmacology , Dendritic Spines/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Dentate Gyrus/drug effects , Disease Models, Animal , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Mice, Inbred DBA , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Synapses/drug effects , Synapses/pathology , Up-Regulation
9.
J Neurosci ; 34(22): 7600-10, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24872564

ABSTRACT

Calcium/calmodulin-dependent protein kinase II (CaMKII) is essential for synaptic plasticity underlying memory formation. Some functions of CaMKII are mediated by interactions with synaptic proteins, and activity-triggered translocation of CaMKII to synapses has been heavily studied. However, CaMKII actions away from the postsynaptic density (PSD) remain poorly understood, in part because of the difficulty in discerning where CaMKII binds in live cells. We used photoactivated localization microscopy (PALM) in rat hippocampal neurons to track single molecules of CaMKIIα, mapping its spatial and kinetic heterogeneity at high resolution. We found that CaMKIIα exhibits at least three kinetic subpopulations, even within individual spines. Latrunculin application or coexpression of CaMKIIß carrying its actin-binding domain strongly modulated CaMKII diffusion, indicating that a major subpopulation is regulated by the actin cytoskeleton. CaMKII in spines was typically more slowly mobile than in dendrites, consistent with presence of a higher density of binding partners or obstacles. Importantly, NMDA receptor stimulation that triggered CaMKII activation prompted the immobilization and presumed binding of CaMKII in spines not only at PSDs but also at other points up to several hundred nanometers away, suggesting that activated kinase does not target only the PSD. Consistent with this, single endogenous activated CaMKII molecules detected via STORM immunocytochemistry were concentrated in spines both at the PSD and at points quite distant from the synapse. Together, these results indicate that CaMKII mobility within spines is determined by association with multiple interacting proteins, even outside the PSD, suggesting diverse mechanisms by which CaMKII may regulate synaptic transmission.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Dendritic Spines/chemistry , Dendritic Spines/enzymology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/analysis , Cells, Cultured , Dendrites/chemistry , Dendrites/enzymology , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/chemistry , Hippocampus/cytology , Hippocampus/enzymology , Male , Microscopy, Confocal/methods , Rats
10.
J Biol Chem ; 289(5): 2600-9, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24352656

ABSTRACT

Development of dendritic spines is important for synaptic function, and alteration in spine morphogenesis is often associated with mental disorders. Rich2 was an uncharacterized Rho-GAP protein. Here we searched for a role of this protein in spine morphogenesis. We found that it is enriched in dendritic spines of cultured hippocampal pyramidal neurons during early stages of development. Rich2 specifically stimulated the Rac1 GTPase in these neurons. Inhibition of Rac1 by EHT 1864 increased the size and decreased the density of dendritic spines. Similarly, Rich2 overexpression increased the size and decreased the density of dendritic spines, whereas knock-down of the protein by specific si-RNA decreased both size and density of spines. The morphological changes were reflected by the increased amplitude and decreased frequency of miniature EPSCs induced by Rich2 overexpression, while si-RNA treatment decreased both amplitude and frequency of these events. Finally, treatment of neurons with EHT 1864 rescued the phenotype induced by Rich2 knock-down. These results suggested that Rich2 controls dendritic spine morphogenesis and function via inhibition of Rac1.


Subject(s)
Dendritic Spines/enzymology , GTPase-Activating Proteins/metabolism , Neurons/enzymology , Neuropeptides/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , COS Cells , Chlorocebus aethiops , Excitatory Postsynaptic Potentials/physiology , GTPase-Activating Proteins/genetics , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/growth & development , Mice , Morphogenesis/physiology , Neurogenesis/physiology , Neurons/ultrastructure , Neuropeptides/genetics , Patch-Clamp Techniques , Primary Cell Culture , rac1 GTP-Binding Protein/genetics
11.
Nature ; 458(7236): 299-304, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19295602

ABSTRACT

Calcium/calmodulin-dependent kinase II (CaMKII) plays a central part in long-term potentiation (LTP), which underlies some forms of learning and memory. Here we monitored the spatiotemporal dynamics of CaMKII activation in individual dendritic spines during LTP using two-photon fluorescence lifetime imaging microscopy, in combination with two-photon glutamate uncaging. Induction of LTP and associated spine enlargement in single spines triggered transient ( approximately 1 min) CaMKII activation restricted to the stimulated spines. CaMKII in spines was specifically activated by NMDA receptors and L-type voltage-sensitive calcium channels, presumably by nanodomain Ca(2+) near the channels, in response to glutamate uncaging and depolarization, respectively. The high degree of compartmentalization and channel specificity of CaMKII signalling allow stimuli-specific spatiotemporal patterns of CaMKII signalling and may be important for synapse-specificity of synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendritic Spines/enzymology , Dendritic Spines/physiology , Long-Term Potentiation/physiology , Animals , Calcium/antagonists & inhibitors , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line , Cells, Cultured , Chelating Agents/pharmacology , Enzyme Activation/drug effects , Fluorescence , Fluorescence Resonance Energy Transfer , Glutamic Acid/metabolism , Hippocampus/cytology , Humans , Kinetics , Photons , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synaptic Potentials/physiology , Time Factors
12.
J Neurosci ; 32(27): 9288-300, 2012 Jul 04.
Article in English | MEDLINE | ID: mdl-22764236

ABSTRACT

Many cellular processes involve a small number of molecules and undergo stochastic fluctuations in their levels of activity. Cerebellar long-term depression (LTD) is a form of synaptic plasticity expressed as a reduction in the number of synaptic AMPA receptors (AMPARs) in Purkinje cells. We developed a stochastic model of the LTD signaling network, including a PKC-ERK-cPLA(2) positive feedback loop and mechanisms of AMPAR trafficking, and tuned the model to replicate calcium uncaging experiments. The signaling network activity in single synapses switches between two discrete stable states (LTD and non-LTD) in a probabilistic manner. The stochasticity of the signaling network causes threshold dithering and allows at the macroscopic level for many different and stable mean magnitudes of depression. The probability of LTD occurrence in a single spine is only modulated by the concentration and duration of the signal used to trigger it, and inputs with the same magnitude can give rise to two different responses; there is no threshold for the input signal. The stochasticity is intrinsic to the signaling network and not mostly dependent on noise in the calcium input signal, as has been suggested previously. The activities of the ultrasensitive ERK and of cPLA(2) undergo strong stochastic fluctuations. Conversely, PKC, which acts as a noise filter, is more constantly activated. Systematic variation of the biochemical population size demonstrates that threshold dithering and the absence of spontaneous LTD depend critically on the number of molecules in a spine, indicating constraints on spine size in Purkinje cells.


Subject(s)
Cerebellum/physiology , Long-Term Synaptic Depression/physiology , MAP Kinase Signaling System/physiology , Models, Neurological , Animals , Calcium/physiology , Cerebellum/cytology , Cerebellum/pathology , Dendritic Spines/enzymology , Dendritic Spines/pathology , Dendritic Spines/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Feedback, Physiological/physiology , Humans , Neural Pathways/enzymology , Neural Pathways/pathology , Neural Pathways/physiology , Neuronal Plasticity/physiology , Phospholipases A2, Cytosolic/physiology , Probability , Protein Kinase C/physiology , Purkinje Cells/enzymology , Purkinje Cells/pathology , Purkinje Cells/physiology , Receptors, AMPA/physiology , Stochastic Processes , Synaptic Transmission/physiology
13.
J Neurosci ; 32(7): 2314-23, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22396406

ABSTRACT

Adolescence is characterized by vulnerability to the development of neuropsychiatric disorders including drug addiction, as well as prefrontal cortical refinement that culminates in structural stability in adulthood. Neuronal refinement and stabilization are hypothesized to confer resilience to poor decision making and addictive-like behaviors, although intracellular mechanisms are largely unknown. We characterized layer V prefrontal dendritic spine development and refinement in adolescent wild-type mice and mice lacking the cytoskeletal regulatory protein Abl-related gene (Arg) kinase. Relative to hippocampal CA1 pyramidal neurons, which exhibited a nearly linear increase in spine density up to postnatal day 60 (P60), wild-type prefrontal spine density peaked at P31, and then declined by 18% by P56-P60. In contrast, dendritic spines in mice lacking Arg destabilized by P31, leading to a net loss in both structures. Destabilization corresponded temporally to the emergence of exaggerated psychomotor sensitivity to cocaine. Moreover, cocaine reduced dendritic spine density in wild-type orbitofrontal cortex and enlarged remaining spine heads, but arg(-/-) spines were unresponsive. Local application of Arg or actin polymerization inhibitors exaggerated cocaine sensitization, as did reduced gene dosage of the Arg substrate, p190RhoGAP. Genetic and pharmacological Arg inhibition also retarded instrumental reversal learning and potentiated responding for reward-related cues, providing evidence that Arg regulates both psychomotor sensitization and decision-making processes implicated in addiction. These findings also indicate that structural refinement in the adolescent orbitofrontal cortex mitigates psychostimulant sensitivity and support the emerging perspective that the structural response to cocaine may, at any age, have behaviorally protective consequences.


Subject(s)
Arginine Kinase/physiology , Cocaine/pharmacology , Dendritic Spines/enzymology , Neuronal Plasticity , Prefrontal Cortex/enzymology , Action Potentials/genetics , Age Factors , Animals , Arginine Kinase/deficiency , Dendritic Spines/drug effects , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neurogenesis/drug effects , Neurogenesis/genetics , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Prefrontal Cortex/drug effects
14.
J Biol Chem ; 287(36): 30084-96, 2012 Aug 31.
Article in English | MEDLINE | ID: mdl-22815483

ABSTRACT

p21-activated kinase 1 (PAK1) and PAK3 belong to group I of the PAK family and control cell movement and division. They also regulate dendritic spine formation and maturation in the brain, and play a role in synaptic transmission and synaptic plasticity. PAK3, in particular, is known for its implication in X-linked intellectual disability. The pak3 gene is expressed in neurons as a GTPase-regulated PAK3a protein and also as three splice variants which display constitutive kinase activity. PAK1 regulation is based on its homodimerization, forming an inactive complex. Here, we analyze the PAK3 capacity to dimerize and show that although PAK3a is able to homodimerize, it is more likely to form heterodimeric complexes with PAK1. We further show that two intellectual disability mutations impair dimerization with PAK1. The b and c inserts present in the regulatory domain of PAK3 splice variants decrease the dimerization but retain the capacity to form heterodimers with PAK1. PAK1 and PAK3 are co-expressed in neurons, are colocalized within dendritic spines, co-purify with post-synaptic densities, and co-immunoprecipitate in brain lysates. Using kinase assays, we demonstrate that PAK1 inhibits the activity of PAK3a but not of the splice variant PAK3b in a trans-regulatory manner. Altogether, these results show that PAK3 and PAK1 signaling may be coordinated by heterodimerization.


Subject(s)
Dendritic Spines/enzymology , Nerve Tissue Proteins/metabolism , Post-Synaptic Density/enzymology , Protein Multimerization , p21-Activated Kinases/metabolism , Alternative Splicing/genetics , Animals , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic/genetics , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , HeLa Cells , Humans , Intellectual Disability/enzymology , Intellectual Disability/genetics , Mice , Mutation , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary , Signal Transduction/genetics , p21-Activated Kinases/genetics
15.
J Biol Chem ; 287(12): 9346-59, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22282498

ABSTRACT

Actin cytoskeletal remodeling plays a critical role in transforming the morphology of subcellular structures across various cell types. In the brain, restructuring of dendritic spines through actin cytoskeleletal reorganization is implicated in the regulation of synaptic efficacy and the storage of information in neural circuits. However, the upstream pathways that provoke actin-based spine changes remain only partly understood. Here we show that EphA receptor signaling remodels spines by triggering a sequence of events involving actin filament rearrangement and synapse/spine reorganization. Rapid EphA signaling over minutes activates the actin filament depolymerizing/severing factor cofilin, alters F-actin distribution in spines, and causes transient spine elongation through the phosphatases slingshot 1 (SSH1) and calcineurin/protein phosphatase 2B (PP2B). This early phase of spine extension is followed by synaptic reorganization events that take place over minutes to hours and involve the relocation of pre/postsynaptic components and ultimately spine retraction. Thus, EphA receptors utilize discrete cellular and molecular pathways to promote actin-based structural plasticity of excitatory synapses.


Subject(s)
Actins/metabolism , Dendritic Spines/enzymology , Dendritic Spines/metabolism , Ephrins/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Actins/genetics , Animals , Calcineurin/genetics , Calcineurin/metabolism , Cells, Cultured , Cofilin 1/genetics , Cofilin 1/metabolism , Dendritic Spines/genetics , Humans , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/genetics , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Spine/cytology , Spine/enzymology , Spine/metabolism , Synapses/metabolism
16.
Dev Cell ; 14(2): 150-2, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18267081

ABSTRACT

Cell polarity is essential for many biological processes and is regulated by conserved protein complexes, including the Par complex, Rho GTPases, and their regulators. In this issue of Developmental Cell, studies by Nakayama et al. and Zhang and Macara examine how interplay between Rho GTPases and the Par complex control polarized cell migration and dendritic spine morphogenesis in alternate ways.


Subject(s)
Carrier Proteins/metabolism , Cell Polarity , rho-Associated Kinases/metabolism , Animals , Carrier Proteins/chemistry , Dendritic Spines/enzymology , Humans , Protein Kinase C/metabolism , Rats , cdc42 GTP-Binding Protein/metabolism
17.
Dev Cell ; 14(2): 216-26, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18267090

ABSTRACT

The majority of excitatory synaptic transmission in the brain occurs at dendritic spines, which are actin-rich protrusions on the dendrites. The asymmetric nature of these structures suggests that proteins regulating cell polarity might be involved in their formation. Indeed, the polarity protein PAR-3 is required for normal spine morphogenesis. However, this function is independent of association with atypical protein kinase C (aPKC) and PAR-6. Here we show that PAR-6 together with aPKC plays a distinct but essential role in spine morphogenesis. Knockdown of PAR-6 inhibits spine morphogenesis, whereas overexpression of PAR-6 increases spine density, and these effects are mediated by aPKC. Using a FRET biosensor, we further show that p190 RhoGAP and RhoA act downstream of the PAR-6/aPKC complex. These results define a role for PAR-6 and aPKC in dendritic spine biogenesis and maintenance, and reveal an unexpected link between the PAR-6/aPKC complex and RhoA activity.


Subject(s)
Carrier Proteins/metabolism , Cell Polarity , DNA-Binding Proteins/metabolism , Dendritic Spines/enzymology , Morphogenesis , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Mutation/genetics , Protein Kinase C/metabolism , Rats , rhoA GTP-Binding Protein/metabolism
18.
PLoS Comput Biol ; 8(2): e1002383, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22346744

ABSTRACT

Dopamine release in the striatum has been implicated in various forms of reward dependent learning. Dopamine leads to production of cAMP and activation of protein kinase A (PKA), which are involved in striatal synaptic plasticity and learning. PKA and its protein targets are not diffusely located throughout the neuron, but are confined to various subcellular compartments by anchoring molecules such as A-Kinase Anchoring Proteins (AKAPs). Experiments have shown that blocking the interaction of PKA with AKAPs disrupts its subcellular location and prevents LTP in the hippocampus and striatum; however, these experiments have not revealed whether the critical function of anchoring is to locate PKA near the cAMP that activates it or near its targets, such as AMPA receptors located in the post-synaptic density. We have developed a large scale stochastic reaction-diffusion model of signaling pathways in a medium spiny projection neuron dendrite with spines, based on published biochemical measurements, to investigate this question and to evaluate whether dopamine signaling exhibits spatial specificity post-synaptically. The model was stimulated with dopamine pulses mimicking those recorded in response to reward. Simulations show that PKA colocalization with adenylate cyclase, either in the spine head or in the dendrite, leads to greater phosphorylation of DARPP-32 Thr34 and AMPA receptor GluA1 Ser845 than when PKA is anchored away from adenylate cyclase. Simulations further demonstrate that though cAMP exhibits a strong spatial gradient, diffusible DARPP-32 facilitates the spread of PKA activity, suggesting that additional inactivation mechanisms are required to produce spatial specificity of PKA activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Spines/enzymology , Models, Neurological , Neuronal Plasticity/physiology , Animals , Computational Biology , Computer Simulation , Cyclic AMP/metabolism , Dopamine/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Humans , Intracellular Space/metabolism , Monte Carlo Method , Reproducibility of Results , Signal Transduction
19.
J Math Biol ; 66(7): 1499-525, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22588358

ABSTRACT

CaMKII (Ca²âº-calmodulin-dependent protein kinase II) is a key regulator of glutamatergic synapses and plays an essential role in many forms of synaptic plasticity. It has recently been observed experimentally that stimulating a local region of dendrite not only induces the local translocation of CaMKII from the dendritic shaft to synaptic targets within spines, but also initiates a wave of CaMKII translocation that spreads distally through the dendrite with an average speed of order 1 µm/s. We have previously developed a simple reaction-diffusion model of CaMKII translocation waves that can account for the observed wavespeed and predicts wave propagation failure if the density of spines is too high. A major simplification of our previous model was to treat the distribution of spines as spatially uniform. However, there are at least two sources of heterogeneity in the spine distribution that occur on two different spatial scales. First, spines are discrete entities that are joined to a dendritic branch via a thin spine neck of submicron radius, resulting in spatial variations in spine density at the micron level. The second source of heterogeneity occurs on a much longer length scale and reflects the experimental observation that there is a slow proximal to distal variation in the density of spines. In this paper, we analyze how both sources of heterogeneity modulate the speed of CaMKII translocation waves along a spiny dendrite. We adapt methods from the study of the spread of biological invasions in heterogeneous environments, including homogenization theory of pulsating fronts and Hamilton-Jacobi dynamics of sharp interfaces.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Dendritic Spines/physiology , Models, Neurological , Protein Transport/physiology , Synapses/physiology , Animals , Computer Simulation , Dendritic Spines/enzymology , Rats
20.
Biochem J ; 444(3): 457-64, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22413754

ABSTRACT

In vitro, the tumour suppressor PTEN (phosphatase and tensin homologue deleted on chromosome 10) displays intrinsic phosphatase activity towards both protein and lipid substrates. In vivo, the lipid phosphatase activity of PTEN, through which it dephosphorylates the 3 position in the inositol sugar of phosphatidylinositol derivatives, is important for its tumour suppressor function; however, the significance of its protein phosphatase activity remains unclear. Using two-photon laser-scanning microscopy and biolistic gene delivery of GFP (green fluorescent protein)-tagged constructs into organotypic hippocampal slice cultures, we have developed an assay of PTEN function in living tissue. Using this bioassay, we have demonstrated that overexpression of wild-type PTEN led to a decrease in spine density in neurons. Furthermore, it was the protein phosphatase activity, but not the lipid phosphatase activity, of PTEN that was essential for this effect. The ability of PTEN to decrease neuronal spine density depended upon the phosphorylation status of serine and threonine residues in its C-terminal segment and the integrity of the C-terminal PDZ-binding motif. The present study reveals a new aspect of the function of this important tumour suppressor and suggest that, in addition to dephosphorylating the 3 position in phosphatidylinositol phospholipids, the critical protein substrate of PTEN may be PTEN itself.


Subject(s)
Dendritic Spines/enzymology , Hippocampus/enzymology , PTEN Phosphohydrolase/physiology , Amino Acid Sequence , Animals , Animals, Newborn , Dendritic Spines/ultrastructure , Enzyme Activation/physiology , Hippocampus/ultrastructure , Molecular Sequence Data , Organ Culture Techniques , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/physiology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL