Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters











Publication year range
1.
J Neurosci ; 41(4): 578-593, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33262245

ABSTRACT

The dynamic regulation of DNA methylation in postmitotic neurons is necessary for memory formation and other adaptive behaviors. Ten-eleven translocation 1 (TET1) plays a part in these processes by oxidizing 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), thereby initiating active DNA demethylation. However, attempts to pinpoint its exact role in the nervous system have been hindered by contradictory findings, perhaps due in part, to a recent discovery that two isoforms of the Tet1 gene are differentially expressed from early development into adulthood. Here, we demonstrate that both the shorter transcript (Tet1S ) encoding an N-terminally truncated TET1 protein and a full-length Tet1 (Tet1FL ) transcript encoding canonical TET1 are co-expressed in the adult mouse brain. We show that Tet1S is the predominantly expressed isoform and is highly enriched in neurons, whereas Tet1FL is generally expressed at lower levels and more abundant in glia, suggesting their roles are at least partially cell type-specific. Using viral-mediated, isoform and neuron-specific molecular tools, we find that the individual repression of each transcript leads to the dysregulation of unique gene ensembles and contrasting changes in basal synaptic transmission. In addition, Tet1S repression enhances, while Tet1FL impairs, hippocampal-dependent memory in male mice. Together, our findings demonstrate that each Tet1 isoform serves a distinct role in the mammalian brain.SIGNIFICANCE STATEMENT In the brain, activity-dependent changes in gene expression are required for the formation of long-term memories. DNA methylation plays an essential role in orchestrating these learning-induced transcriptional programs by influencing chromatin accessibility and transcription factor binding. Once thought of as a stable epigenetic mark, DNA methylation is now known to be impermanent and dynamically regulated, driving neuroplasticity in the brain. We found that Tet1, a member of the ten-eleven translocation (TET) family of enzymes that mediates removal of DNA methyl marks, is expressed as two separate isoforms in the adult mouse brain and that each differentially regulates gene expression, synaptic transmission and memory formation. Together, our findings demonstrate that each Tet1 isoform serves a distinct role in the CNS.


Subject(s)
Brain/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation/genetics , Memory/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Animals , Anxiety/genetics , Anxiety/psychology , Conditioning, Classical , Epigenesis, Genetic/physiology , Fear/psychology , Hippocampus/physiology , Isomerism , Male , Mice , Mice, Inbred C57BL , Neuroglia/physiology , Neurons/physiology
2.
Neurobiol Aging ; 32(12): 2198-210, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20189687

ABSTRACT

The transcription of genes that support memory processes are likely to be impacted by the normal aging process. Because Arc is necessary for memory consolidation and enduring synaptic plasticity, we examined Arc transcription within the aged hippocampus. Here, we report that Arc transcription is reduced within the aged hippocampus compared to the adult hippocampus during both "off line" periods of rest, and following spatial behavior. This reduction is observed within ensembles of CA1 "place cells", which make less mRNA per cell, and in the dentate gyrus (DG) where fewer granule cells are activated by behavior. In addition, we present data suggesting that aberrant changes in methylation of the Arc gene may be responsible for age-related decreases in Arc transcription within CA1 and the DG. Given that Arc is necessary for normal memory function, these subregion-specific epigenetic and transcriptional changes may result in less efficient memory storage and retrieval during aging.


Subject(s)
Aging/genetics , Cytoskeletal Proteins/biosynthesis , DNA Methylation/genetics , Hippocampus/metabolism , Nerve Tissue Proteins/biosynthesis , Transcription, Genetic/physiology , Aging/pathology , Animals , Base Sequence , Cytoskeletal Proteins/genetics , Hippocampus/pathology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Rats , Rats, Inbred F344 , Spatial Behavior/physiology
3.
Cell Mol Life Sci ; 63(9): 1009-16, 2006 May.
Article in English | MEDLINE | ID: mdl-16596331

ABSTRACT

In this review we address the idea that conservation of epigenetic mechanisms for information storage represents a unifying model in biology, with epigenetic mechanisms being utilized for cellular memory at levels from behavioral memory to development to cellular differentiation. Epigenetic mechanisms typically involve alterations in chromatin structure, which in turn regulate gene expression. An emerging idea is that the regulation of chromatin structure through histone acetylation and DNA methylation may mediate long-lasting behavioral change in the context of learning and memory. We find this idea fascinating because similar mechanisms are used for triggering and storing long-term 'memory' at the cellular level, for example when cells differentiate. An additional intriguing aspect of the hypothesis of a role for epigenetic mechanisms in information storage is that lifelong behavioral memory storage may involve lasting changes in the physical, three-dimensional structure of DNA itself.


Subject(s)
Epigenesis, Genetic , Memory/physiology , Models, Neurological , Neuronal Plasticity/genetics , Animals , Chromatin/genetics , Hippocampus/metabolism , Nervous System/metabolism
4.
Neuroscience ; 133(4): 969-81, 2005.
Article in English | MEDLINE | ID: mdl-15916859

ABSTRACT

The transcription factor nuclear factor-kappa B (NF-kappaB) is an inducible regulator of genes that plays a crucial role in the nervous system. Glutamate receptor stimulation is one well-described mechanism for NF-kappaB activation. In the studies presented here we used the glutamate analog, kainate to investigate the signaling mechanisms that couple to NF-kappaB activation in hippocampus. Kainate (250 nM) application to hippocampal slices elicited a time-dependent increase in nuclear NF-kappaB levels in areas CA3 and CA1, but not dentate, compared with controls. Further analysis focused on hippocampal area CA3, revealed increased NF-kappaB DNA binding activity in response to kainate stimulation. Supershift electrophoretic mobility shift assay indicated that the kainate-mediated NF-kappaB complex binding DNA was composed of p65, p50, and c-Rel subunits. Through inhibition studies we found that extracellular signal-regulated protein kinase (ERK) and phosphatidylinositol-3 kinase (PI3K) couple to basal and kainate-mediated NF-kappaB DNA binding activity in area CA3. Kainate elicited decreased total and increased phospho-inhibitor kappa B alpha (IkappaBalpha), suggesting that kainate-mediated activation of NF-kappaB is via the classical IkappaB kinase pathway. Interestingly, inhibition of ERK but not PI3K blocked the kainate-mediated increase in phospho-IkappaBalpha. Thus, our findings support a role for the ERK and PI3K pathways in kainate-mediated NF-kappaB activation in hippocampal area CA3, but these kinases may target the NF-kappaB pathway at different loci.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Hippocampus/drug effects , Kainic Acid/pharmacology , MAP Kinase Kinase Kinases/physiology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/physiology , Animals , Blotting, Western/methods , Chromones/pharmacology , Drug Interactions , Electrophoretic Mobility Shift Assay/methods , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Immunohistochemistry/methods , In Vitro Techniques , Lamin Type B/metabolism , Male , Morpholines/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Time Factors
5.
Science ; 306(5697): 882-4, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15514161

ABSTRACT

The prefrontal cortex is a higher brain region that regulates thought, behavior, and emotion using representational knowledge, operations often referred to as working memory. We tested the influence of protein kinase C (PKC) intracellular signaling on prefrontal cortical cognitive function and showed that high levels of PKC activity in prefrontal cortex, as seen for example during stress exposure, markedly impair behavioral and electrophysiological measures of working memory. These data suggest that excessive PKC activation can disrupt prefrontal cortical regulation of behavior and thought, possibly contributing to signs of prefrontal cortical dysfunction such as distractibility, impaired judgment, impulsivity, and thought disorder.


Subject(s)
Memory/physiology , Prefrontal Cortex/physiology , Protein Kinase C/metabolism , Adrenergic alpha-Agonists/pharmacology , Alkaloids , Animals , Benzophenanthridines , Carbolines/pharmacology , Electrophysiology , Enzyme Activation , Female , Imidazoles/pharmacology , Lithium Carbonate/pharmacology , Macaca mulatta , Male , Memory/drug effects , Neurons/drug effects , Neurons/physiology , Phenanthridines/pharmacology , Prefrontal Cortex/enzymology , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/physiology , Signal Transduction , Stress, Physiological/physiopathology , Tetradecanoylphorbol Acetate/pharmacology , Valproic Acid/pharmacology
6.
Neuroscience ; 126(2): 305-12, 2004.
Article in English | MEDLINE | ID: mdl-15207348

ABSTRACT

Alzheimer's disease is a learning and memory disorder pathologically characterized by the deposition of beta-amyloid plaques and loss of neurons and synapses in affected areas of the brain. Mutations in presenilin 1 (PS1) lead to the most aggressive form of familial Alzheimer's disease (FAD), and are associated with accelerated plaque deposition. However, since the function of PS1 is pleiotropic, we reasoned that the FAD mutations may alter multiple PS1-mediated pathways, and the combination of which may account for the early onset nature of the disease phenotype. Using the PS1M146V knockin mice in which the M146V mutation was incorporated into the endogenous mouse PS1 gene, we report here that the FAD mutation results in impaired hippocampus-dependent associative learning, as measured by a contextual fear conditioning paradigm, at 3 months of age. This is correlated with reduced adult neurogenesis in the dentate gyrus. However, short-term and long-term synaptic plasticity in both area CA1 and dentate gyrus are not affected. Our results suggest that impaired adult neurogenesis may contribute to the memory deficit associated with FAD.


Subject(s)
Association Learning/physiology , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Membrane Proteins/genetics , Mutation/genetics , Animals , Conditioning, Psychological/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Presenilin-1
7.
Neuron ; 31(5): 671-4, 2001 Sep 13.
Article in English | MEDLINE | ID: mdl-11567607

ABSTRACT

Studies of the signal transduction mechanisms underlying learning and memory have provided many new insights into the molecular mechanisms underlying associative conditioning in mammals. In this issue of Neuron, Gean and colleagues report the discovery that the PI-3 kinase/AKT(PKB) pathway contributes to LTP and the consolidation of amygdala-dependent cued fear conditioning in rats.


Subject(s)
Amygdala/physiology , Memory/physiology , Proto-Oncogenes/physiology , Animals , Conditioning, Psychological/physiology , Humans , Long-Term Potentiation/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology
9.
J Neurosci ; 21(12): 4125-33, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11404397

ABSTRACT

Alzheimer's Disease (AD) is the most common of the senile dementias, the prevalence of which is increasing rapidly, with a projected 14 million affected worldwide by 2025. The signal transduction mechanisms that underlie the learning and memory derangements in AD are poorly understood. beta-Amyloid (Abeta) peptides are elevated in brain tissue of AD patients and are the principal component of amyloid plaques, a major criterion for postmortem diagnosis of the disease. Using acute and organotypic hippocampal slice preparations, we demonstrate that Abeta peptide 1-42 (Abeta42) couples to the mitogen-activated protein kinase (MAPK) cascade via alpha7 nicotinic acetylcholine receptors (nAChRs). In vivo elevation of Abeta, such as that exhibited in an animal model for AD, leads to the upregulation of alpha7 nAChR protein. alpha7 nAChR upregulation occurs concomitantly with the downregulation of the 42 kDa isoform of extracellular signal-regulated kinase (ERK2) MAPK in hippocampi of aged animals. The phosphorylation state of a transcriptional mediator of long-term potentiation and a downstream target of the ERK MAPK cascade, the cAMP-regulatory element binding (CREB) protein, were affected also. These findings support the model that derangement of hippocampus signal transduction cascades in AD arises as a consequence of increased Abeta burden and chronic activation of the ERK MAPK cascade in an alpha7 nAChR-dependent manner that eventually leads to the downregulation of ERK2 MAPK and decreased phosphorylation of CREB protein.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Hippocampus/metabolism , MAP Kinase Signaling System/drug effects , Peptide Fragments/pharmacology , Receptors, Nicotinic/metabolism , Aging/metabolism , Alzheimer Disease/etiology , Animals , Cells, Cultured , Chronic Disease , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Heterozygote , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Maze Learning/drug effects , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Nicotine/pharmacology , Nicotinic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Up-Regulation , alpha7 Nicotinic Acetylcholine Receptor
10.
Curr Biol ; 11(10): R391-4, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11378403

ABSTRACT

Protein phosphorylation has long been known to play a key role in triggering the synaptic changes underlying learning and memory. Recent studies highlight the importance of tightly regulated dephosphorylation as a mechanism controlling the induction of long-term synaptic change and lasting memory.


Subject(s)
Memory/physiology , Proteins/metabolism , Phosphorylation , Proteins/physiology , Synapses/metabolism , Synapses/physiology
11.
J Neurochem ; 77(4): 961-71, 2001 May.
Article in English | MEDLINE | ID: mdl-11359861

ABSTRACT

Hippocampal long-term potentiation (LTP) is a robust and long-lasting form of synaptic plasticity that is the leading candidate for a cellular mechanism contributing to mammalian learning and memory. Investigations over the past decade have revealed that the biochemistry of LTP induction involves mechanisms of great subtlety and complexity. This review highlights themes that have emerged as a result of our increased knowledge of the signal transduction pathways involved in the induction of NMDA receptor-dependent LTP in area CA1 of the hippocampus. Among these themes are signal amplification, signal integration and signal coordination. Here we use these themes as an organizing context for reviewing the profusion of signaling mechanisms involved in the induction of LTP.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Animals , Calcium/physiology , Feedback , Hippocampus/metabolism , Models, Neurological , Receptors, AMPA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Signal Transduction
12.
J Neurosci ; 21(10): 3383-91, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11331368

ABSTRACT

Changes in gene expression are thought to be involved in neuronal plasticity associated with learning and memory. Although acetylation of lysine residues on histones by histone acetyltransferases (HAT) is an obligatory component of transcription, HAT activity has been largely ignored in studies of the nervous system. We developed a new model for studying novel taste learning using novel solid food presentation to nondeprived animals. Using this behavioral paradigm, we investigated short- and long-term regulation of lysine acetyltransferase activity and the ERK/mitogen-activated protein kinase (MAPK)/RSK cascade in insular cortex, a CNS region known to be crucial for the formation of novel taste memories. We observed that novel taste learning elicited biphasic (acute and long-lasting) activation of two distinct lysine acetyltransferase activities along with the ERK/MAPK cascade in insular cortex. In vitro studies revealed that the ERK cascade could regulate the lysine acetylation of a 42 kDa lysine acetyltransferase substrate, suggesting a causal relationship between ERK activation and lysine acetyltransferase activity in insular cortex. Overall, our studies reveal an unanticipated long-lasting activation of insular cortex signal transduction cascades in novel taste learning. Furthermore, our studies suggest the hypothesis that acute and long-term ERK activation and lysine-histone acetyltransferase activation may play a role in regulating gene expression in single-trial learning and long-term memory formation.


Subject(s)
Acetyltransferases/metabolism , Cerebral Cortex/enzymology , Discrimination Learning/physiology , MAP Kinase Signaling System/physiology , Saccharomyces cerevisiae Proteins , Taste/physiology , Acetylation/drug effects , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cerebral Cortex/chemistry , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Discrimination Learning/drug effects , Eating/drug effects , Eating/physiology , Enzyme Inhibitors/pharmacology , Histone Acetyltransferases , Histone Deacetylase Inhibitors , In Vitro Techniques , Lithium Chloride/pharmacology , Lysine/metabolism , Male , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Protein Kinases/metabolism , Ribosomal Protein S6 Kinases , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Mol Psychiatry ; 6(2): 246-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11317232

ABSTRACT

Preliminary clinical data indicate that omega-3 fatty acids may be effective mood stabilizers for patients with bipolar disorder. Both lithium and valproic acid are known to inhibit protein kinase C (PKC) activity after subchronic administration in cell culture and in vivo. The current study was undertaken to determine the effects of the omega-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on protein kinase C phosphotransferase activity in vitro. Various concentrations of DHA, EPA, and arachidonic acid (AA) were incubated with the catalytic domain of protein kinase C beta from rat brain. Protein kinase C activity was measured by quantifying incorporation of (32)P-PO(4) into a synthetic peptide substrate. Both DHA and EPA, as well as the combination of DHA and EPA, inhibited PKC activity at concentrations as low as 10 micromol l(-1). In contrast, arachidonic acid had no effect on PKC activity. Thus, PKC represents a potential site of action of omega-3 fatty acids in their effects on the treatment of bipolar disorder.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Protein Kinase C/metabolism , Animals , Arachidonic Acid/pharmacology , Arachidonic Acids/pharmacology , Bipolar Disorder/drug therapy , Bipolar Disorder/enzymology , Brain/enzymology , Docosahexaenoic Acids/pharmacology , Enzyme Activation/drug effects , In Vitro Techniques , Rats
14.
Rev Neurosci ; 12(1): 41-50, 2001.
Article in English | MEDLINE | ID: mdl-11236064

ABSTRACT

We address in this review the various types of chemical reactions that underlie memory storage in biological systems. Using examples from both invertebrate and mammalian learning systems, we describe three types of memory-storing reactions: short-term reactions mediated by transient changes in second messenger levels, long-term reactions mediated by species with long half lives, and ultralong-term or mnemogenic reactions that can store memory indefinitely, even in the face of ongoing turnover of the molecules involved.


Subject(s)
Central Nervous System/metabolism , Memory/physiology , Neurons/metabolism , Animals , Humans , Models, Animal , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Second Messenger Systems/physiology , Synaptic Transmission/physiology
15.
Learn Mem ; 8(1): 11-9, 2001.
Article in English | MEDLINE | ID: mdl-11160759

ABSTRACT

The extracellular signal-regulated kinases (ERKs) are members of the mitogen-activated protein kinase (MAPK) superfamily of enzymes and have recently garnered considerable attention in the field of learning and memory. ERK activation has been shown to be required for the induction of long-term potentiation (LTP) in the rat hippocampus and for the formation of associative and spatial memories in both the rat and the mouse. However, the individual roles for the two isoforms of ERK have yet to be deciphered. To investigate the specific contribution of the ERK1 (p44) isoform of MAPK to mammalian learning, we performed a general behavioral and physiological characterization of mice lacking the ERK1 gene. The ERK1-null animals demonstrated significantly higher levels of activity in the open field test. However, we observed no other discernible deficits in the ERK1 knockout mice in our behavioral testing. Specifically, no differences were observed in the acquisition or retention (24 h and 2 wk after training) of either contextual or cue fear conditioning between the ERK1(-/-) and their wild-type littermate controls. In addition, no learning phenotype was observed in the passive avoidance test. When hippocampal slices were analyzed, we found no deficits in baseline synaptic transmission or in tetanus-induced LTP in hippocampal area CA1. We found no apparent compensatory changes in the expression of ERK2 (p42 MAPK). We conclude that hippocampus- and amygdala-dependent emotional learning does not depend critically on the activity of ERK1.


Subject(s)
Emotions/physiology , Learning/physiology , Mitogen-Activated Protein Kinases/physiology , Animals , Hippocampus/physiology , Mice , Mice, Knockout/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Motor Activity/physiology , Reference Values , Sensation/physiology
16.
J Neurochem ; 76(1): 1-10, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145972

ABSTRACT

The mitogen-activated protein kinase (MAP kinase, MAPK) cascade, as the name implies, was originally discovered as a critical regulator of cell division and differentiation. As further details of this signaling cascade were worked out, it became clear that the MAPK cascade is in fact a prototype for a family of signaling cascades that share the motif of three serially linked kinases regulating each other by sequential phosphorylation. Thus, a revised nomenclature arose that uses the term MAPK to refer to the entire superfamily of signaling cascades (comprising the erks, the JNKs and the p38 stress activated protein kinases), and specifies the prototype MAPK as the extracellular signal-regulated kinase (erk). The two erk MAPK isoforms, p44 MAPK and p42 MAPK, are referred to as erk1 and erk2, respectively. The erks are abundantly expressed in neurons in the mature central nervous system, raising the question of why the prototype molecular regulators of cell division and differentiation are present in these non-dividing, terminally differentiated neurons. This review will describe the beginnings of an answer to this question. Interestingly, the general model has begun to emerge that the erk signaling system has been co-opted in mature neurons to function in synaptic plasticity and memory. Moreover, recent insights have led to the intriguing prospect that these molecules serve as biochemical signal integrators and molecular coincidence detectors for coordinating responses to extracellular signals in neurons. In this review I will first outline the essential components of this signal transduction cascade, and briefly describe recent results implicating the erks in mammalian synaptic plasticity and learning. I will then proceed to outline recent results implicating the erks as molecular signal integrators and, potentially, coincidence detectors. Finally, I will speculate on what the critical downstream effectors of the erks are in neurons, and how they might provide a readout of the integrated signal.


Subject(s)
MAP Kinase Signaling System/physiology , Memory/physiology , Neuronal Plasticity/physiology , Neurons/enzymology , Potassium Channels, Voltage-Gated , Signal Transduction/physiology , Animals , Avoidance Learning/physiology , Cell Membrane/metabolism , Conditioning, Psychological , Cyclic AMP Response Element-Binding Protein/metabolism , Fear , Hippocampus/cytology , Hippocampus/metabolism , Humans , Long-Term Potentiation/physiology , Maze Learning/physiology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Potassium Channels/metabolism , Rats , Shal Potassium Channels , Synaptic Transmission/physiology
17.
J Biol Chem ; 276(14): 10888-96, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11152679

ABSTRACT

Recent data suggest that omega-3 fatty acids may be effective in epilepsy, cardiovascular disorders, arthritis, and as mood stabilizers for bipolar disorder; however, the mechanism of action of these compounds is unknown. Based on earlier studies implicating omega-3 fatty acids as inhibitors of protein kinase C activity in intact cells, we hypothesized that omega-3 fatty acids may act through direct inhibition of second messenger-regulated kinases and sought to determine whether the omega-3 double bond might uniquely confer pharmacologic efficacy and potency for fatty acids of this type. In our studies we observed that omega-3 fatty acids inhibited the in vitro activities of cAMP-dependent protein kinase, protein kinase C, Ca(2+)/calmodulin-dependent protein kinase II, and the mitogen-activated protein kinase (MAPK). Our results with a series of long-chain fatty acid structural homologs suggest an important role for the omega-3 double bond in conferring inhibitory efficacy. To assess whether omega-3 fatty acids were capable of inhibiting protein kinases in living neurons, we evaluated their effect on signal transduction pathways in the hippocampus. We found that omega-3 fatty acids could prevent serotonin receptor-induced MAPK activation in hippocampal slice preparations. In addition, we evaluated the effect of omega-3 fatty acids on hippocampal long-term potentiation, a form of synaptic plasticity known to be dependent on protein kinase activation. We observed that omega-3 fatty acids blocked long-term potentiation induction without inhibiting basal synaptic transmission. Overall, our results from both in vitro and live cell preparations suggest that inhibition of second messenger-regulated protein kinases is one locus of action of omega-3 fatty acids.


Subject(s)
Fatty Acids, Omega-3/metabolism , Hippocampus/metabolism , Protein Kinases/metabolism , Signal Transduction , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Omega-3/pharmacology , Rats , Signal Transduction/drug effects
18.
J Neurochem ; 75(6): 2277-87, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080179

ABSTRACT

The mitogen-activated protein kinase ERK has recently become a focus of studies of synaptic plasticity and learning and memory. Due to the prominent role of potassium channels in regulating the electrical properties of membranes, modulation of these channels by ERK could play an important role in mediating learning-related synaptic plasticity in the CNS. Kv4.2 is a Shal-type potassium channel that passes an A-type current and is localized to dendrites and cell bodies in the hippocampus. The sequence of Kv4.2 contains several consensus sites for ERK phosphorylation. In the present studies, we tested the hypothesis that Kv4.2 is an ERK substrate. We determined that the Kv4.2 C-terminal cytoplasmic domain is an effective ERK2 substrate, and that it is phosphorylated at three sites: Thr(602), Thr(607), and Ser(616). We used this information to develop antibodies that recognize Kv4.2 phosphorylated by ERK2. One of our phospho-site-selective antibodies was generated using a triply phosphorylated peptide as the antigen. We determined that this antibody recognizes ERK-phosphorylated Kv4.2 in COS-7 cells transfected with Kv4.2 and native ERK-phosphorylated Kv4.2 in the rat hippocampus. These observations indicate that Kv4.2 is a substrate for ERK in vitro and in vivo, and suggest that ERK may regulate potassium-channel function by direct phosphorylation of the pore-forming alpha subunit.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Amino Acid Sequence , Animals , Antibodies/metabolism , Antibody Specificity , Binding Sites/drug effects , Blotting, Western , COS Cells , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , In Vitro Techniques , Mitogen-Activated Protein Kinase 1/pharmacology , Molecular Sequence Data , Phosphorylation/drug effects , Potassium Channels/chemistry , Protein Structure, Tertiary/drug effects , Rats , Shal Potassium Channels
19.
Learn Mem ; 7(5): 321-32, 2000.
Article in English | MEDLINE | ID: mdl-11040264

ABSTRACT

Voltage-gated A-type potassium channels such as Kv4.2 regulate generation of action potentials and are localized abundantly in the hippocampus and striatum. Phosphorylation consensus sites for various kinases exist within the sequence of the potassium channel subunit Kv4.2, including consensus sites for extracellular signal-regulated kinase/mitogen activated protein kinase (ERK/MAPK), protein kinase A (PKA), protein kinase C (PKC), and calcium/calmodulin-dependent kinase II (CaMKII), and kinase assays have shown that particular amino acids of the consensus sites are bonafide phosphorylation sites in vitro. We have developed antibodies recognizing Kv4.2 triply phosphorylated at the three ERK sites as well as two antibodies recognizing singly phosphorylated Kv4.2 channels at the PKA sites (one amino-terminal and one carboxy-terminal). In the present study, we report the development of reliable immunohistochemistry protocols to study the localization of these phosphorylated versions of Kv4.2, as well as total Kv4.2 in the mouse brain. A general description of the areas highlighted by these antibodies includes the hippocampus, amygdala, cortex, and cerebellum. Such areas display robust synaptic plasticity and have been implicated in spatial, associative, and motor learning. Interestingly, in the hippocampus, the antibodies to differentially phosphorylated Kv4.2 channels localize to specific afferent pathways, indicating that the Kv4.2 phosphorylation state may be input specific. For example, the stratum lacunosum moleculare, which receives inputs from the entorhinal cortex via the perforant pathway, displays relatively little ERK-phosphorylated Kv4.2 or PKA carboxy-terminal-phosphorylated Kv4.2. However, this same layer is highlighted by antibodies that recognize Kv4.2 that has been phosphorylated by PKA at the amino terminus. Similarly, of the three antibodies tested, the soma of CA3 neurons are primarily recognized by the ERK triply phosphorylated Kv4.2 antibody, and the mossy fiber inputs to CA3 are primarily recognized by the carboxy-terminal PKA-phosphorylated Kv4.2. This differential phosphorylation is particularly interesting in two contexts. First, phosphorylation may be serving as a mechanism for targeting. For example, the amino-terminal PKA phosphorylation may be acting as a tag for a discrete pool of Kv4.2 to enter stratum lacunosum moleculare. Second, as phosphorylation may regulate channel biophysical properties, differential phosphorylation of Kv4.2 in the dendrites of pyramidal neurons may confer unique biophysical properties upon particular dendritic input layers.


Subject(s)
Brain/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Amygdala/metabolism , Animals , Cerebellum/metabolism , Hippocampus/metabolism , Immunohistochemistry , Mice , Phosphorylation , Protein Isoforms/metabolism , Shal Potassium Channels , Somatosensory Cortex/metabolism , Tissue Distribution
20.
Acta Neurobiol Exp (Wars) ; 60(3): 377-94, 2000.
Article in English | MEDLINE | ID: mdl-11016081

ABSTRACT

Long-term potentiation (LTP), a cellular model for long-term memory, is generally acknowledged to consist of both a short-term phase that is characterized by a dependence on autonomous protein kinase activity, and a long-term phase that is characterized by a dependence on changes in gene expression and new protein synthesis. Similarly, long-term memory exhibits a dependence on gene expression and altered protein synthesis. Recent evidence indicates that the mitogen-activated protein kinase (MAPK) cascade plays a role in both LTP and long-term memory. The MAPK cascade has heretofore largely been studied in the context of cell division and proliferation and as such, mechanisms for the regulation of gene expression by the MAPK cascade have received considerable attention. Given the possible role of altered gene expression in the late phase of LTP and in long-term memory, we evaluated the capacity of the MAPK ERK (extracellular signal-regulated kinase) to regulate phosphorylation of the transcription factor cAMP response element binding protein (CREB) in hippocampal area CA1. Our studies indicate a critical role for the MAPK cascade in the regulation of CREB phosphorylation in the hippocampus.


Subject(s)
Central Nervous System/enzymology , Gene Expression Regulation, Enzymologic/genetics , Mitogen-Activated Protein Kinases/biosynthesis , Mitogen-Activated Protein Kinases/genetics , Animals , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Long-Term Potentiation
SELECTION OF CITATIONS
SEARCH DETAIL