Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Neurosci ; 35(19): 7503-8, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972176

ABSTRACT

Increasing plasticity in neurons of the prefrontal cortex (PFC) has been proposed as a possible therapeutic tool to enhance extinction, a process that is impaired in post-traumatic stress disorder, schizophrenia, and addiction. To test this hypothesis, we generated transgenic mice that overexpress neurogranin (a calmodulin-binding protein that facilitates long-term potentiation) in the PFC. Neurogranin overexpression in the PFC enhanced long-term potentiation and increased the rates of extinction learning of both fear conditioning and sucrose self-administration. Our results indicate that elevated neurogranin function within the PFC can enhance local plasticity and increase the rate of extinction learning across different behavioral tasks. Thus, neurogranin can provide a molecular link between enhanced plasticity and enhanced extinction.


Subject(s)
Extinction, Psychological/physiology , Neurogranin/metabolism , Neuronal Plasticity/genetics , Prefrontal Cortex/physiology , Analysis of Variance , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Conditioning, Classical/physiology , Conditioning, Operant/physiology , Electric Stimulation , Fear/physiology , In Vitro Techniques , Long-Term Potentiation/genetics , Male , Mice , Mice, Transgenic , Neurogranin/genetics , Prefrontal Cortex/cytology , Pyramidal Cells/metabolism , Sucrose/administration & dosage
2.
EMBO J ; 28(19): 3027-39, 2009 Oct 07.
Article in English | MEDLINE | ID: mdl-19713936

ABSTRACT

Learning-correlated plasticity at CA1 hippocampal excitatory synapses is dependent on neuronal activity and NMDA receptor (NMDAR) activation. However, the molecular mechanisms that transduce plasticity stimuli to postsynaptic potentiation are poorly understood. Here, we report that neurogranin (Ng), a neuron-specific and postsynaptic protein, enhances postsynaptic sensitivity and increases synaptic strength in an activity- and NMDAR-dependent manner. In addition, Ng-mediated potentiation of synaptic transmission mimics and occludes long-term potentiation (LTP). Expression of Ng mutants that lack the ability to bind to, or dissociate from, calmodulin (CaM) fails to potentiate synaptic transmission, strongly suggesting that regulated Ng-CaM binding is necessary for Ng-mediated potentiation. Moreover, knocking-down Ng blocked LTP induction. Thus, Ng-CaM interaction can provide a mechanistic link between induction and expression of postsynaptic potentiation.


Subject(s)
Calmodulin/metabolism , Neurogranin/metabolism , Neuronal Plasticity , Neurons/cytology , Synaptic Transmission , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/genetics , Cells, Cultured , Gene Expression , Hippocampus/cytology , Long-Term Potentiation , Neurogranin/analysis , Neurogranin/genetics , Protein Binding , Rats , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spine/ultrastructure
3.
J Cell Biol ; 222(12)2023 12 04.
Article in English | MEDLINE | ID: mdl-37938213

ABSTRACT

Mutations in IQSEC2/BRAG1 cause intellectual dysfunction by impairing ARF-GEF activity and long-term depression. In this issue, Bai et al. (https://doi.org/10.1083/jcb.202307117) discover how constitutive ARF-GEF activity is regulated by a closed conformation which opens in the presence of Ca2+. Two known pathogenic mutations cause "leaky" autoinhibition with reduced synaptic dynamic range and impaired cognitive performance.


Subject(s)
Guanine Nucleotide Exchange Factors , Neuronal Plasticity , Mutation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Calcium , Cognition
4.
Mol Pain ; 8: 62, 2012 Sep 10.
Article in English | MEDLINE | ID: mdl-22963123

ABSTRACT

BACKGROUND: Sickle cell disease (SCD) is associated with both acute vaso-occlusive painful events as well as chronic pain syndromes, including heightened sensitivity to touch. We have previously shown that mice with severe SCD (HbSS mice; express 100% human sickle hemoglobin in red blood cells; RBCs) have sensitized nociceptors, which contribute to increased mechanical sensitivity. Yet, the hypersensitivity in these neural populations alone may not fully explain the mechanical allodynia phenotype in mouse and humans. FINDINGS: Using the Light Touch Behavioral Assay, we found HbSS mice exhibited increased responses to repeated application of both innocuous punctate and dynamic force compared to control HbAA mice (100% normal human hemoglobin). HbSS mice exhibited a 2-fold increase in percent response to a 0.7mN von Frey monofilament when compared to control HbAA mice. Moreover, HbSS mice exhibited a 1.7-fold increase in percent response to the dynamic light touch "puffed" cotton swab stimulus. We further investigated the mechanisms that drive this behavioral phenotype by focusing on the cutaneous sensory neurons that primarily transduce innocuous, light touch. Low threshold cutaneous afferents from HbSS mice exhibited sensitization to mechanical stimuli that manifested as an increase in the number of evoked action potentials to suprathreshold force. Rapidly adapting (RA) AƟ and AƎĀ“ D-hair fibers showed the greatest sensitization, each with a 75% increase in suprathreshold firing compared to controls. Slowly adapting (SA) AƟ afferents had a 25% increase in suprathreshold firing compared to HbAA controls. CONCLUSIONS: These novel findings demonstrate mice with severe SCD exhibit mechanical allodynia to both punctate and dynamic light touch and suggest that this behavioral phenotype may be mediated in part by the sensitization of light touch cutaneous afferent fibers to suprathreshold force. These findings indicate that AƟ fibers can be sensitized to mechanical force and should potentially be examined for sensitization in other tissue injury and disease models.


Subject(s)
Anemia, Sickle Cell/complications , Anemia, Sickle Cell/pathology , Hyperalgesia/complications , Hyperalgesia/pathology , Mechanoreceptors/metabolism , Skin/pathology , Touch , Action Potentials , Anemia, Sickle Cell/physiopathology , Animals , Humans , Hyperalgesia/physiopathology , Mice , Motor Activity , Nerve Fibers/metabolism , Nerve Fibers/pathology , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Physical Stimulation , Skin/metabolism , Skin/physiopathology
5.
Am J Physiol Heart Circ Physiol ; 302(6): H1285-93, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22245774

ABSTRACT

Recent studies have indicated that inhibitors of the synthesis of 20-hydroxyeicosatetraenoic acid (20-HETE) may have direct neuroprotective actions since they reduce infarct volume after ischemia reperfusion in the brain without altering blood flow. To explore this possibility, the present study used organotypic hippocampal slice cultures subjected to oxygen-glucose deprivation (OGD) and reoxygenation to examine whether 20-HETE is released by organotypic hippocampal slices after OGD and whether it contributes to neuronal death through the generation of ROS and activation of caspase-3. The production of 20-HETE increased twofold after OGD and reoxygenation. Blockade of the synthesis of 20-HETE with N-hydroxy-N'-(4-butyl-2-methylphenol)formamidine (HET0016) or its actions with a 20-HETE antagonist, 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid, reduced cell death, as measured by the release of lactate dehydrogenase and propidium iodide uptake. Administration of a 20-HETE mimetic, 20-hydroxyeicosa-5(Z),14(Z)-dienoic acid (5,14-20-HEDE), had the opposite effect and increased injury after OGD. The death of neurons after OGD was associated with an increase in the production of ROS and activation of caspase-3. These effects were attenuated by HET0016 and potentiated after the administration of 5,14-20-HEDE. These findings indicate that the production of 20-HETE by hippocampal slices is increased after OGD and that inhibitors of the synthesis or actions of 20-HETE protect neurons from ischemic cell death. The protective effect of 20-HETE inhibitors is associated with a decrease in superoxide production and activation of caspase-3.


Subject(s)
Amidines/pharmacology , Glucose/deficiency , Hippocampus/drug effects , Hydroxyeicosatetraenoic Acids/antagonists & inhibitors , Hydroxyeicosatetraenoic Acids/pharmacology , Neuroprotective Agents/pharmacology , Animals , Animals, Newborn , Caspase 3/metabolism , Cell Death/drug effects , Cell Hypoxia , Cytoprotection , Hippocampus/metabolism , Hippocampus/pathology , Hydroxyeicosatetraenoic Acids/metabolism , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Time Factors , Tissue Culture Techniques
6.
Eur J Neurosci ; 33(2): 244-50, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21198977

ABSTRACT

Learning-related potentiation of synaptic strength at Cornu ammonis subfield 1 (CA1) hippocampal excitatory synapses is dependent on neuronal activity and the activation of glutamate receptors. However, molecular mechanisms that regulate and fine-tune the expression of long-term potentiation (LTP) are not well understood. Recently it has been indicated that neurogranin (Ng), a neuron-specific, postsynaptic protein that is phosphorylated by protein kinase C, potentiates synaptic transmission in an LTP-like manner. Here, we report that a Ng mutant that is unable to be phosphorylated cannot potentiate synaptic transmission in rat CA1 hippocampal neurons and results in a submaximal expression of LTP. Our results provide the first evidence that the phosphorylation of Ng can regulate LTP expression.


Subject(s)
Long-Term Potentiation/physiology , Neurogranin/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Phosphorylation , Rats , Synapses/metabolism , Synaptic Transmission/physiology
7.
Small GTPases ; 11(1): 1-7, 2020 01.
Article in English | MEDLINE | ID: mdl-29363391

ABSTRACT

Precise trafficking events, such as those that underlie synaptic transmission and plasticity, require complex regulation. G-protein signaling plays an essential role in the regulation of membrane and protein trafficking. However, it is not well understood how small GTPases and their regulatory proteins coordinate such specific events. Our recent publication focused on a highly abundant synaptic GEF, BRAG1, whose physiologic relevance was unknown. We find that BRAG1s GEF activity is required for activity-dependent trafficking of AMPARs. Moreover, BRAG1 bidirectionally regulates synaptic transmission in a manner independent of this activity. In addition to the GEF domain, BRAG1 contains several functional domains whose roles are not yet understood but may mediate protein-protein interactions and regulatory effects necessary for its role in regulation of AMPAR trafficking. In this commentary, we explore the potential for BRAG1 to provide specificity of small GTPase signaling, coordinating activity-dependent activation of small GTPase activity with signaling and scaffolding molecules involved in trafficking through its GEF activity and other functional domains.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Monomeric GTP-Binding Proteins/metabolism , Animals , Biological Transport , Humans
8.
Hippocampus ; 19(1): 66-78, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18680156

ABSTRACT

Hypothyroidism induces cognitive impairment in experimental animals and patients. Clinical reports are conflicting about the ability of thyroid hormone replacement therapy to fully restore the hypothyroidism-induced learning and memory impairment. In this study, we investigated the effects of L-thyroxin (thyroxin) treatment on hippocampus-dependent learning and memory in thyroidectomized adult rats. In the radial arm water maze (RAWM) task, thyroxin treated thyroidectomized animals made significantly fewer errors than the untreated hypothyroid animals in Trial 3 of the acquisition phase, short-term memory and long-term memory tests. In addition, the number of errors made by the thyroxin treated thyroidectomized animals was not different from that of the control group. Furthermore, the days-to-criterion (DTC) values for thyroxin treated thyroidectomized animals were not different from those of the control group but significantly lower than those of the untreated hypothyroid animals. In anesthetized rats, extracellular recording from hippocampal area CA1 of hypothyroid rats shows that thyroxin treatment restores impaired Late-phase long-term potentiation (L-LTP). Immunoblot analysis of signaling molecules, including cyclic-AMP response element binding protein (CREB), mitogen-activated protein kinases (MAPKp44/42; ERK1/2), in area CA1 revealed that thyroxin treatment reversed hypothyroidism-induced reduction of signaling molecules essential for learning and memory, and L-LTP. This study shows that thyroxin treatment reverses hypothyroidism-induced impairment of hippocampus-dependent cognition, and L-LTP, probably by restoring the levels of signaling molecule important for these processes.


Subject(s)
Hippocampus/drug effects , Hypothyroidism/complications , Hypothyroidism/drug therapy , Memory Disorders/drug therapy , Memory Disorders/etiology , Thyroxine/pharmacology , Animals , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Hypothyroidism/metabolism , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Memory Disorders/metabolism , Rats , Rats, Wistar , Thyroxine/therapeutic use , Treatment Outcome
9.
Front Mol Neurosci ; 12: 322, 2019.
Article in English | MEDLINE | ID: mdl-32038160

ABSTRACT

Long-term potentiation (LTP) and long-term depression (LTD) are two major forms of synaptic plasticity that are widely accepted as cellular mechanisms involved in learning and memory. Metaplasticity is a process whereby modifications in synaptic processes shift the threshold for subsequent plasticity. While metaplasticity has been functionally observed, its molecular basis is not well understood. Here, we report that neurogranin (Ng) regulates metaplasticity by shifting the threshold toward potentiation, i.e., increasing Ng in hippocampal neurons lowers the threshold for LTP and augments the threshold for LTD. We also show that Ng does not change the ultrastructural localization of calmodulin (CaM)-dependent protein Kinase II (CaMKII) or calcineurin, critical enzymes for the induction of LTP and LTD, respectively. Interestingly, while CaMKII concentrates close to the plasma membrane, calcineurin concentrates away from the plasma membrane. These data, along with the previous observation showing Ng targets CaM closer to the plasma membrane, suggesting that shifting the localization of CaM within the dendritic spines and closer to the plasma membrane, where there is more CaMKII, may be favoring the activation of CaMKII vs. that of calcineurin. Thus, the regulation of CaM localization/targeting within dendritic spines by Ng may provide a mechanistic basis for the regulation of metaplasticity.

10.
Front Mol Neurosci ; 12: 43, 2019.
Article in English | MEDLINE | ID: mdl-30842726

ABSTRACT

We have recently described an A350V mutation in IQSEC2 associated with intellectual disability, autism and epilepsy. We sought to understand the molecular pathophysiology of this mutation with the goal of developing targets for drug intervention. We demonstrate here that the A350V mutation results in interference with the binding of apocalmodulin to the IQ domain of IQSEC2. We further demonstrate that this mutation results in constitutive activation of the guanine nucleotide exchange factor (GEF) activity of IQSEC2 resulting in increased production of the active form of Arf6. In a CRISPR generated mouse model of the A350V IQSEC2 mutation, we demonstrate that the surface expression of GluA2 AMPA receptors in mouse hippocampal tissue was significantly reduced in A350V IQSEC2 mutant mice compared to wild type IQSEC2 mice and that there is a significant reduction in basal synaptic transmission in the hippocampus of A350V IQSEC2 mice compared to wild type IQSEC2 mice. Finally, the A350V IQSEC2 mice demonstrated increased activity, abnormal social behavior and learning as compared to wild type IQSEC2 mice. These findings suggest a model of how the A350V mutation in IQSEC2 may mediate disease with implications for targets for drug therapy. These studies provide a paradigm for a personalized approach to precision therapy for a disease that heretofore has no therapy.

11.
Exp Neurol ; 277: 115-123, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26721336

ABSTRACT

Amyloid Ɵ (AƟ) is widely considered one of the early causes of cognitive deficits observed in Alzheimer's disease. Many of the deficits caused by AƟ are attributed to its disruption of synaptic function represented by its blockade of long-term potentiation (LTP) and its induction of synaptic depression. Identifying pathways that reverse these synaptic deficits may open the door to new therapeutic targets. In this study, we explored the possibility that Neurogranin (Ng)-a postsynaptic calmodulin (CaM) targeting protein that enhances synaptic function-may rescue AƟ-mediated deficits in synaptic function. Our results show that Ng is able to reverse synaptic depression and LTP deficits induced by AƟ. Furthermore, Ng's restoration of synaptic transmission is through the insertion of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPARs). These restorative effects of Ng are dependent on the interaction of Ng and CaM and CaM-dependent activation of CaMKII. Overall, this study identifies a novel mechanism to rescue synaptic deficits induced by AƟ oligomers. It also suggests Ng and CaM signaling as potential therapeutic targets for Alzheimer's disease as well as important tools to further explore the pathophysiology underlying the disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Hippocampus/cytology , Long-Term Potentiation/drug effects , Neurogranin/pharmacology , Neurons/drug effects , Synaptic Transmission/drug effects , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Animals, Newborn , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Humans , In Vitro Techniques , Mutagenesis , Mutation/genetics , Nerve Net/drug effects , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
12.
Nat Commun ; 7: 11080, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27009485

ABSTRACT

Dysfunction of the proteins regulating synaptic function can cause synaptic plasticity imbalance that underlies neurological disorders such as intellectual disability. A study found that four distinct mutations within BRAG1, an Arf-GEF synaptic protein, each led to X-chromosome-linked intellectual disability (XLID). Although the physiological functions of BRAG1 are poorly understood, each of these mutations reduces BRAG1's Arf-GEF activity. Here we show that BRAG1 is required for the activity-dependent removal of AMPA receptors in rat hippocampal pyramidal neurons. Moreover, we show that BRAG1 bidirectionally regulates synaptic transmission. On one hand, BRAG1 is required for the maintenance of synaptic transmission. On the other hand, BRAG1 expression enhances synaptic transmission, independently of BRAG1 Arf-GEF activity or neuronal activity, but dependently on its C-terminus interactions. This study demonstrates a dual role of BRAG1 in synaptic function and highlights the functional relevance of reduced BRAG1 Arf-GEF activity as seen in the XLID-associated human mutations.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Long-Term Synaptic Depression , Synaptic Transmission , Amino Acid Sequence , Guanine Nucleotide Exchange Factors/chemistry , HEK293 Cells , Humans , Receptors, AMPA/metabolism
13.
Nat Neurosci ; 19(3): 443-53, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26780512

ABSTRACT

Dyshomeostasis of amyloid-Ɵ peptide (AƟ) is responsible for synaptic malfunctions leading to cognitive deficits ranging from mild impairment to full-blown dementia in Alzheimer's disease. AƟ appears to skew synaptic plasticity events toward depression. We found that inhibition of PTEN, a lipid phosphatase that is essential to long-term depression, rescued normal synaptic function and cognition in cellular and animal models of Alzheimer's disease. Conversely, transgenic mice that overexpressed PTEN displayed synaptic depression that mimicked and occluded AƟ-induced depression. Mechanistically, AƟ triggers a PDZ-dependent recruitment of PTEN into the postsynaptic compartment. Using a PTEN knock-in mouse lacking the PDZ motif, and a cell-permeable interfering peptide, we found that this mechanism is crucial for AƟ-induced synaptic toxicity and cognitive dysfunction. Our results provide fundamental information on the molecular mechanisms of AƟ-induced synaptic malfunction and may offer new mechanism-based therapeutic targets to counteract downstream AƟ signaling.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Cognition Disorders/physiopathology , PTEN Phosphohydrolase/physiology , Synaptic Transmission/physiology , Alzheimer Disease/complications , Amyloid beta-Peptides/toxicity , Animals , Cognition Disorders/complications , Disease Models, Animal , Gene Knock-In Techniques , Mice , Mice, Transgenic , PDZ Domains/genetics , PDZ Domains/physiology , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Primary Cell Culture , Rats , Synaptic Transmission/drug effects
14.
J Neurosci ; 24(20): 4758-66, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15152036

ABSTRACT

The delivery of neurotransmitter receptors into synapses is essential for synaptic function and plasticity. In particular, AMPA-type glutamate receptors (AMPA receptors) reach excitatory synapses according to two distinct routes: a regulated pathway, which operates transiently during synaptic plasticity, and a constitutive pathway, which maintains synaptic function under conditions of basal transmission. However, the specific mechanisms that distinguish these two trafficking pathways are essentially unknown. Here, we evaluate the role of the molecular chaperone hsp90 (heat shock protein 90) in excitatory synaptic transmission in the hippocampus. On one hand, we found that hsp90 is necessary for the efficient neurotransmitter release at the presynaptic terminal. In addition, we identified hsp90 as a critical component of the cellular machinery that delivers AMPA receptors into the postsynaptic membrane. Using the hsp90-specific inhibitors radicicol and geldanamycin, we show that hsp90 is required for the constitutive trafficking of AMPA receptors into synapses during their continuous cycling between synaptic and nonsynaptic sites. In contrast, hsp90 function is not required for either the surface delivery of AMPA receptors into the nonsynaptic plasma membrane or for the acute, regulated delivery of AMPA receptors into synapses during plasticity induction (long-term potentiation). The synaptic cycling of AMPA receptors was also blocked by an hsp90-binding tetratricopeptide repeat (TPR) domain, suggesting that the role of hsp90 in AMPA receptor trafficking is mediated by a TPR domain-containing protein. These results demonstrate new roles for hsp90 in synaptic function by controlling neurotransmitter release and, independently, by mediating the continuous cycling of synaptic AMPA receptors.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , Neurotransmitter Agents/metabolism , Pyramidal Cells/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Vesicular Transport Proteins , Adenosine Triphosphatases/physiology , Animals , Carrier Proteins/physiology , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Hippocampus/cytology , Hippocampus/physiology , In Vitro Techniques , Long-Term Potentiation/physiology , N-Ethylmaleimide-Sensitive Proteins , Patch-Clamp Techniques , Protein Structure, Tertiary/physiology , Protein Transport/drug effects , Protein Transport/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid/physiology , Signal Transduction/physiology , Sindbis Virus/genetics , Synaptic Transmission/physiology
15.
Methods Enzymol ; 403: 153-66, 2005.
Article in English | MEDLINE | ID: mdl-16473584

ABSTRACT

Members of the Rab family of small GTPases are essential regulators of intracellular membrane sorting. Nevertheless, very little is known about the role of these proteins in the membrane trafficking processes that operate at synapses, and specifically, at postsynaptic terminals. These events include the activity-dependent exocytic and endocytic trafficking of AMPA-type glutamate receptors, which underlies long-lasting forms of synaptic plasticity such as long-term potentiation (LTP) and long-term depression (LTD). This chapter summarizes different experimental methods to address the role of Rab proteins in the trafficking of neurotransmitter receptors at postsynaptic terminals in the hippocampus. These techniques include immunogold electron microscopy to ultrastructurally localize endogenous Rab proteins at synapses, molecular biology methods to express recombinant Rab proteins in hippocampal slice cultures, electrophysiological techniques to evaluate the role of Rab proteins in synaptic transmission, and confocal fluorescence imaging to monitor receptor trafficking at dendrites and spines and its dependence on Rab proteins.


Subject(s)
Hippocampus/metabolism , Receptors, Neurotransmitter/physiology , Synapses/metabolism , rab GTP-Binding Proteins/physiology , Animals , Hippocampus/enzymology , Hippocampus/physiology , Synapses/enzymology , Synapses/physiology
16.
Sci Rep ; 5: 11135, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26084473

ABSTRACT

Calmodulin (CaM) plays a key role in synaptic function and plasticity due to its ability to mediate Ca(2+) signaling. Therefore, it is essential to understand the dynamics of CaM at dendritic spines. In this study we have explored CaM dynamics using live-cell confocal microscopy and fluorescence recovery after photobleaching (FRAP) to study CaM diffusion. We find that only a small fraction of CaM in dendritic spines is immobile. Furthermore, the diffusion rate of CaM was regulated by neurogranin (Ng), a CaM-binding protein enriched at dendritic spines. Interestingly, Ng did not influence the immobile fraction of CaM at recovery plateau. We have previously shown that Ng enhances synaptic strength in a CaM-dependent manner. Taken together, these data indicate that Ng-mediated enhancement of synaptic strength is due to its ability to target, rather than sequester, CaM within dendritic spines.


Subject(s)
Calmodulin/metabolism , Dendritic Spines/metabolism , Neurogranin/metabolism , Animals , Calcium Signaling , Calmodulin/genetics , Gene Expression , Hippocampus/metabolism , Microscopy, Confocal , Neurogranin/genetics , Phosphorylation , Protein Binding , Rats , Synapses/metabolism
17.
Behav Brain Res ; 155(1): 77-84, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15325781

ABSTRACT

Both hypothyroidism and stress interfere with cognitive function in patients. This study examined the effect of hypothyroidism and stress on hippocampus-dependent learning and memory in rats using the novel radial arm water maze (RAWM), which measures spatial working memory. Hypothyroidism was accomplished by thyroidectomy and 2 weeks later a form of intruder stress was used as the chronic psychosocial stressor. After 4-6 weeks of stress, rats were trained to learn (during the acquisition phase; four trials) and then remember (during two memory test trials occurring 15 and 120 min after the acquisition phase) the within-day location of a hidden escape platform, which was in different arm every day. The number of errors (entry into arms other than the platform arm) was noted. Within-day learning of the platform location was largely unaffected by the experimental manipulations, indicating that rats in all groups were equally capable of finding the platform to escape from the water with similar numbers of errors (P > 0.005). The number of days a rat took to reach a criterion (DTC; a maximum of one error in three consecutive days) indicated that chronic stress or hypothyroidism, alone, resulted in a mild impairment of spatial memory, and the combination of chronic stress and hypothyroidism resulted in a more severe and long-lasting memory impairment. The data indicated that the combination of stress and hypothyroidism produced more deleterious effects on hippocampal function than either chronic stress or hypothyroidism alone.


Subject(s)
Hippocampus/physiology , Hypothyroidism/complications , Memory Disorders/etiology , Stress, Psychological/complications , Animals , Behavior, Animal , Escape Reaction/physiology , Male , Maze Learning/physiology , Rats , Rats, Wistar , Thyroidectomy/methods , Time Factors
18.
J Vis Exp ; (74)2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23609099

ABSTRACT

Immunoelectron microscopy is a powerful tool to study biological molecules at the subcellular level. Antibodies coupled to electron-dense markers such as colloidal gold can reveal the localization and distribution of specific antigens in various tissues. The two most widely used techniques are pre-embedding and post-embedding techniques. In pre-embedding immunogold-electron microscopy (EM) techniques, the tissue must be permeabilized to allow antibody penetration before it is embedded. These techniques are ideal for preserving structures but poor penetration of the antibody (often only the first few micrometers) is a considerable drawback. The post-embedding labeling methods can avoid this problem because labeling takes place on sections of fixed tissues where antigens are more easily accessible. Over the years, a number of modifications have improved the post-embedding methods to enhance immunoreactivity and to preserve ultrastructure. Tissue fixation is a crucial part of EM studies. Fixatives chemically crosslink the macromolecules to lock the tissue structures in place. The choice of fixative affects not only structural preservation but also antigenicity and contrast. Osmium tetroxide (OsO4), formaldehyde, and glutaraldehyde have been the standard fixatives for decades, including for central nervous system (CNS) tissues that are especially prone to structural damage during chemical and physical processing. Unfortunately, OsO4 is highly reactive and has been shown to mask antigens, resulting in poor and insufficient labeling. Alternative approaches to avoid chemical fixation include freezing the tissues. But these techniques are difficult to perform and require expensive instrumentation. To address some of these problems and to improve CNS tissue labeling, Phend et al. replaced OsO4 with uranyl acetate (UA) and tannic acid (TA), and successfully introduced additional modifications to improve the sensitivity of antigen detection and structural preservation in brain and spinal cord tissues. We have adopted this osmium-free post-embedding method to rat brain tissue and optimized the immunogold labeling technique to detect and study synaptic proteins. We present here a method to determine the ultrastructural localization of synaptic proteins in rat hippocampal CA1 pyramidal neurons. We use organotypic hippocampal cultured slices. These slices maintain the trisynaptic circuitry of the hippocampus, and thus are especially useful for studying synaptic plasticity, a mechanism widely thought to underlie learning and memory. Organotypic hippocampal slices from postnatal day 5 and 6 mouse/rat pups can be prepared as described previously), and are especially useful to acutely knockdown or overexpress exogenous proteins. We have previously used this protocol to characterize neurogranin (Ng), a neuron-specific protein with a critical role in regulating synaptic function . We have also used it to characterize the ultrastructural localization of calmodulin (CaM) and Ca(2+)/CaM-dependent protein kinase II (CaMKII). As illustrated in the results, this protocol allows good ultrastructural preservation of dendritic spines and efficient labeling of Ng to help characterize its distribution in the spine. Furthermore, the procedure described here can have wide applicability in studying many other proteins involved in neuronal functions.


Subject(s)
CA1 Region, Hippocampal/chemistry , Immunohistochemistry/methods , Nerve Tissue Proteins/analysis , Synapses/chemistry , Tissue Fixation/methods , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/ultrastructure , Neurons/chemistry , Neurons/metabolism , Neurons/ultrastructure , Rats , Synapses/metabolism , Synapses/ultrastructure
19.
Sci Rep ; 3: 1392, 2013.
Article in English | MEDLINE | ID: mdl-23462742

ABSTRACT

Neuromodulin (Nm) and neurogranin (Ng) are neuron-specific substrates of protein kinase C (PKC). Their interactions with Calmodulin (CaM) are crucial for learning and memory formation in neurons. Here, we report the structure of IQ peptides (24aa) of Nm/Ng complexed with CaM and their functional studies with full-length proteins. Nm/Ng and their respective IQ peptides are intrinsically unstructured; however, upon binding with CaM, IQ motifs adopt a helical conformation. Ser41 (Ser36) of Nm (Ng) is located in a negatively charged pocket in the apo CaM and, when phosphorylated, it will repel Nm/Ng from CaM. These observations explain the mechanism by which PKC-induced Ser phosphorylation blocks the association of Nm/Ng with CaM and interrupts several learning- and memory-associated functions. Moreover, the present study identified Arg as a key CaM interacting residue from Nm/Ng. This residue is crucial for CaM-mediated function, as evidenced by the inability of the Ng mutant (Arg-to-Ala) to potentiate synaptic transmission in CA1 hippocampal neurons.


Subject(s)
Calmodulin/metabolism , GAP-43 Protein/chemistry , Neurogranin/chemistry , Neurons/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , GAP-43 Protein/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Neurogranin/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary , Protein Unfolding , Rats , Sequence Alignment , Synaptic Transmission
20.
PLoS One ; 7(7): e41275, 2012.
Article in English | MEDLINE | ID: mdl-22848456

ABSTRACT

Calcium entry and the subsequent activation of CaMKII trigger synaptic plasticity in many brain regions. The induction of long-term potentiation (LTP) in the CA1 region of the hippocampus requires a relatively high amount of calcium-calmodulin. This requirement is usually explained, based on in vitro and theoretical studies, by the low affinity of CaMKII for calmodulin. An untested hypothesis, however, is that calmodulin is not randomly distributed within the spine and its targeting within the spine regulates LTP. We have previously shown that overexpression of neurogranin enhances synaptic strength in a calmodulin-dependent manner. Here, using post-embedding immunogold labeling, we show that calmodulin is not randomly distributed, but spatially organized in the spine. Moreover, neurogranin regulates calmodulin distribution such that its overexpression concentrates calmodulin closer to the plasma membrane, where a high level of CaMKII immunogold labeling is also found. Interestingly, the targeting of calmodulin by neurogranin results in lowering the threshold for LTP induction. These findings highlight the significance of calmodulin targeting within the spine in synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Calmodulin/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Neurogranin/metabolism , Animals , Cell Membrane/metabolism , Hippocampus/cytology , Rats , Rats, Sprague-Dawley , Spine/cytology , Spine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL