Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Behav Neurosci ; 135(4): 462-468, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33734729

ABSTRACT

Genetic variants in large conductance voltage and calcium sensitive potassium (BKCa) channels have associations with neurodevelopmental disorders such as autism spectrum disorder, fragile X syndrome, and intellectual disability. In the case of fragile X syndrome, early preclinical studies suggest that BKCa channels may be a promising treatment target for neurodevelopmental disorders. While BKCa channel dysfunction has been investigated within the context of fragile X syndrome, it is unknown whether interference with BKCa channel function is inductive for deficits in behavioral domains relevant to neurodevelopmental disorders. This represents a critical gap in our knowledge regarding the relationship between BKCa dysfunction and neurodevelopmental disorders. To explore this concept, we used the BKCa channel antagonist paxilline to evaluate the role of BKCa channel function in phenotypes of neurodevelopmental disorders. Here we used adult male C57BL/6J mice and a series of behavioral paradigms which assessed anxiety-like behavior, locomotor activity, social behavior, and repetitive self-grooming. We found that acute inhibition with paxilline induced a specific social deficit, but not anxiety-like behavior, or hyperactivity. These findings demonstrate proof-of-concept regarding a relationship between BKCa channel impairment and social behavior. Although this is a limited characterization of the BKCa channel in autistic-like behaviors, it provides evidence for this link. Future studies which examine the effective dose range of paxilline and exhaustive assays of behavior relevant to neurodevelopmental disorders will be needed to delineate the parametric space of the paxilline effect, particularly during critical periods of development, and its potential for therapeutic use. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Inhibition, Psychological , Male , Mice , Mice, Inbred C57BL , Phenotype
2.
FASEB J ; 33(2): 2563-2573, 2019 02.
Article in English | MEDLINE | ID: mdl-30303736

ABSTRACT

In humans, vitamin B12 deficiency causes peripheral and CNS manifestations. Loss of myelin in the peripheral nerves and the spinal cord (SC) contributes to peripheral neuropathy and motor deficits. The metabolic basis for the demyelination and brain disorder is unknown. The transcobalamin receptor-knockout mouse ( Cd320-/-) develops cobalamin (Cbl) deficiency in the nervous system, with mild anemia. A decreased S-adenosylmethionine: S-adenosylhomocysteine ratio and increased methionine were seen in the brain with no significant changes in neurotransmitter metabolites. The structural pathology in the SC presented as loss of myelin in the axonal tracts with inflammation. The sciatic nerve (SN) showed increased nonuniform, internodal segments suggesting demyelination, and remyelination in progress. Consistent with these changes, the Cd320-/- mouse showed an increased latency to thermal nociception. Further, lower amplitude of compound action potential in the SN suggested that the functional capacity of the heavily myelinated axons were preferentially compromised, leading to loss of peripheral sensation. Although the metabolic basis for the demyelination and the structural and functional alterations of the nervous system in Cbl deficiency remain unresolved, the Cd320-/- mouse provides a unique model to investigate the pathologic consequences of vitamin B12 deficiency. -Arora, K., Sequeira, J. M., Alarcon, J. M., Wasek, B., Arning, E., Bottiglieri, T., Quadros, E. V. Neuropathology of vitamin B12 deficiency in the Cd320-/- mouse.


Subject(s)
Brain/pathology , Nervous System Diseases/pathology , Nociception , Receptors, Cell Surface/physiology , Vitamin B 12 Deficiency/complications , Animals , Brain/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Neurotransmitter Agents/metabolism , Vitamin B 12 Deficiency/physiopathology
3.
PLoS One ; 12(5): e0177156, 2017.
Article in English | MEDLINE | ID: mdl-28545069

ABSTRACT

Vitamin B12 (cobalamin) deficiency is prevalent worldwide and causes megaloblastic anemia and neurologic deficits. While the anemia can be treated, the neurologic deficits can become refractive to treatment as the disease progresses. Therefore, timely intervention is critical for a favorable outcome. Moreover, the metabolic basis for the neuro-pathologic changes and the role of cobalamin deficiency in the pathology still remains unexplained. Using a transcobalamin receptor / CD320 knockout mouse that lacks the receptor for cellular uptake of transcobalamin bound cobalamin, we aimed to determine whether cobalamin deficiency in the central nervous system produced functional neurologic deficits in the mouse that would parallel those observed in humans. Our behavioral analyses indicate elevated anxiety and deficits in learning, memory and set-shifting of a spatial memory task in the KO mouse. Consistent with the behavioral deficits, the knockout mouse shows impaired expression of the early phase of hippocampal long-term potentiation along with reduced expression of GluR1, decreased brain mass and a significant reduction in the size of nuclei of the hippocampal pyramidal neurons. Our study suggests that the CD320 knockout mouse develops behavioral deficits associated with cobalamin deficiency and therefore could provide a model to understand the metabolic and genetic basis of neuro-pathologic changes due to cobalamin deficiency.


Subject(s)
Hippocampus/pathology , Receptors, Cell Surface/genetics , Vitamin B 12 Deficiency/genetics , Animals , Anxiety Disorders/genetics , Avoidance Learning , Behavior, Animal , Learning , Memory/physiology , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Pyramidal Cells/pathology , Receptors, AMPA/metabolism
4.
Commun Integr Biol ; 8(2): e1017163, 2015.
Article in English | MEDLINE | ID: mdl-26479998

ABSTRACT

Widely thought to be a housekeeping process, the regulation and synthesis of rRNA emerges as a potentially central mechanism for the maintenance of synaptic plasticity and memory. We have recently shown that an essential component of late-phase synaptic plasticity is rRNA biosynthesis - the rate-limiting step in the production of new ribosomes. We hypothesize that a particular population of ribosomes is generated upon learning-associated neural activity to alter the rate of synthesis of plasticity factors at tagged synapses that will support the maintenance of synaptic plasticity and memory.

5.
Front Behav Neurosci ; 8: 292, 2014.
Article in English | MEDLINE | ID: mdl-25221487

ABSTRACT

The hippocampal CA1 field processes spatial information, but the relative importance of intra- vs. extra-hippocampal sources of input into CA1 for spatial behavior is unclear. To characterize the relative roles of these two sources of input, originating in the hippocampal field CA3 and in the medial entorhinal cortex (MEC), we studied effects of discrete neurotoxic lesions of CA3 or MEC on concurrent spatial and nonspatial navigation tasks, and on synaptic transmission in afferents to CA1. Lesions in CA3 or MEC regions that abolished CA3-CA1, or reduced MEC-CA1 synaptic transmission, respectively, impaired spatial navigation and unexpectedly interfered with cue response, suggesting that in certain conditions of training regimen, hippocampal activity may influence behavior otherwise supported by nonhippocampal neural networks. MEC lesions had milder and temporary behavioral effects, but also markedly amplified transmission in the CA3-CA1 pathway. Extensive behavioral training had a similar, but more modest effect on CA3-CA1 transmission. Thus, cortical input to the hippocampus modulates CA1 activity both directly and indirectly, through heterosynaptic interaction, to control information flow in the hippocampal loop. Following damage to hippocampal cortical input, the functional coupling of separate intra- and extra-hippocampal inputs to CA1 involved in normal learning may initiate processes that support recovery of behavioral function. Such a process may explain how CA3 lesions, which do not significantly modify the basic features of CA1 neural activity, nonetheless impair spatial recall, whereas lesions of EC input to CA1, which reduce the spatial selectivity of CA1 firing in foraging rats, have only mild effects on spatial navigation.

6.
PLoS One ; 9(8): e104364, 2014.
Article in English | MEDLINE | ID: mdl-25089620

ABSTRACT

Long-term memory (LTM) formation requires new protein synthesis and new gene expression. Based on our work in Aplysia, we hypothesized that the rRNA genes, stimulation-dependent targets of the enzyme Poly(ADP-ribose) polymerase-1 (PARP-1), are primary effectors of the activity-dependent changes in synaptic function that maintain synaptic plasticity and memory. Using electrophysiology, immunohistochemistry, pharmacology and molecular biology techniques, we show here, for the first time, that the maintenance of forskolin-induced late-phase long-term potentiation (L-LTP) in mouse hippocampal slices requires nucleolar integrity and the expression of new rRNAs. The activity-dependent upregulation of rRNA, as well as L-LTP expression, are poly(ADP-ribosyl)ation (PAR) dependent and accompanied by an increase in nuclear PARP-1 and Poly(ADP) ribose molecules (pADPr) after forskolin stimulation. The upregulation of PARP-1 and pADPr is regulated by Protein kinase A (PKA) and extracellular signal-regulated kinase (ERK)--two kinases strongly associated with long-term plasticity and learning and memory. Selective inhibition of RNA Polymerase I (Pol I), responsible for the synthesis of precursor rRNA, results in the segmentation of nucleoli, the exclusion of PARP-1 from functional nucleolar compartments and disrupted L-LTP maintenance. Taken as a whole, these results suggest that new rRNAs (28S, 18S, and 5.8S ribosomal components)--hence, new ribosomes and nucleoli integrity--are required for the maintenance of long-term synaptic plasticity. This provides a mechanistic link between stimulation-dependent gene expression and the new protein synthesis known to be required for memory consolidation.


Subject(s)
Long-Term Potentiation/genetics , Memory, Long-Term/physiology , Neuronal Plasticity/genetics , Poly(ADP-ribose) Polymerases/biosynthesis , Synapses/genetics , Animals , Colforsin/administration & dosage , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Neuronal Plasticity/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly Adenosine Diphosphate Ribose/biosynthesis , Poly Adenosine Diphosphate Ribose/genetics , Poly(ADP-ribose) Polymerases/genetics , RNA, Ribosomal, 28S/biosynthesis , RNA, Ribosomal, 28S/genetics , Synapses/physiology
7.
J Neurosci ; 30(10): 3813-25, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20220016

ABSTRACT

Reference memory characterizes the long-term storage of information acquired through numerous trials. In contrast, working memory represents the short-term acquisition of trial-unique information. A number of studies in the rodent hippocampus have focused on the contribution of long-term synaptic potentiation (LTP) to long-term reference memory. In contrast, little is known about the synaptic plasticity correlates of hippocampal-based components of working memory. Here, we described a mouse with selective expression of a dominant-negative mutant of the regulatory subunit of protein kinase A (PKA) only in two regions of the hippocampus, the dentate gyrus and area CA1. This mouse showed a deficit in several forms of LTP in both hippocampal subregions and a lowered threshold for the consolidation of long-term synaptic depression (LTD). When trained with one trial per day in a water maze task, mutant mice displayed a deficit in consolidation of long-term memory. In contrast, these mice proved to be more flexible after a transfer test and also showed a delay-dependent increased performance in working memory, when repetitive information (proactive interference) was presented. We suggest that through its bidirectional control over synaptic plasticity PKA can regulate opposing forms of memory. The defect in L-LTP disrupts long-term memory consolidation. The persistence of LTD may allow acquisition of new information by restricting the body of previously stored information and suppressing interference.


Subject(s)
Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/physiology , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , Mutation/physiology , Neural Pathways/physiology , Time Factors
8.
Neurosci Biobehav Rev ; 32(4): 831-51, 2008.
Article in English | MEDLINE | ID: mdl-18281094

ABSTRACT

A decade ago, the synaptic tagging hypothesis was proposed to explain how newly synthesized plasticity products can be specifically targeted to active synapses. A growing number of studies have validated the seminal findings that gave rise to this model, as well as contributed to unveil and expand the range of mechanisms underlying late-associativity and neuronal computation. Here, we will review what it was learnt during this past decade regarding the cellular and molecular mechanisms underlying synaptic tagging and synaptic capture. The accumulated experimental evidence has widened the theoretical framework set by the synaptic tagging and capture (STC) model and introduced concepts that were originally considered part of alternative models for explaining synapse-specific long-term potentiation (LTP). As a result, we believe that the STC model, now improved and expanded with these new ideas and concepts, still represents the most compelling hypothesis to explain late-associativity in synapse-specific plasticity processes. We will also discuss the impact of this model in our view of the integrative capability of neurons and associative learning.


Subject(s)
Neuronal Plasticity/physiology , Synapses/physiology , Animals , Computer Simulation/history , History, 20th Century , History, 21st Century , Models, Neurological , Protein Kinases/metabolism
9.
J Neurosci ; 27(50): 13909-18, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18077703

ABSTRACT

To investigate the role of CREB-mediated gene expression on the excitability of CA1 pyramidal neurons, we obtained intracellular recordings from pyramidal neurons of transgenic mice expressing a constitutively active form of CREB, VP16-CREB, in a regulated and restricted manner. We found that transgene expression increased the neuronal excitability and inhibited the slow and medium afterhyperpolarization currents. These changes may contribute to the reduced threshold for LTP observed in these mice. When strong transgene expression was turned on for prolonged period of time, these mice also showed a significant loss of hippocampal neurons and sporadic epileptic seizures. These deleterious effects were dose dependent and could be halted, but not reversed by turning off transgene expression. Our experiments reveal a new role for hippocampal CREB-mediated gene expression, identify the slow afterhyperpolarization as a primary target of CREB action, provide a new mouse model to investigate temporal lobe epilepsy and associated neurodegeneration, and illustrate the risks of cell death associated to a sustained manipulation of this pathway. As a result, our study has important implications for both the understanding of the cellular bases of learning and memory and the consideration of therapies targeted to the CREB pathway.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Epilepsy, Temporal Lobe/physiopathology , Gene Expression Regulation , Hippocampus/physiopathology , Neurodegenerative Diseases/physiopathology , Pyramidal Cells/physiopathology , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Disease Models, Animal , Electric Stimulation , Epilepsy, Temporal Lobe/genetics , Female , Handling, Psychological , Hippocampus/pathology , Long-Term Potentiation/genetics , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/genetics , Organ Culture Techniques , Patch-Clamp Techniques , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Survival Rate , Synaptic Transmission/genetics
10.
J Neurosci ; 26(1): 256-64, 2006 Jan 04.
Article in English | MEDLINE | ID: mdl-16399695

ABSTRACT

Studies in the rodent hippocampus have demonstrated that when the late phase of long-term potentiation (L-LTP) is induced in a set of synapses by suprathreshold stimulation, L-LTP can also be expressed by other synapses receiving subthreshold stimulation, a phenomenon usually referred as "capture of L-LTP." Because the pyramidal neurons in the mammalian hippocampus have both apical and basal dendrites, we have now investigated whether capture of L-LTP, previously described only within the apical dendritic compartment, can also take place within the basilar dendritic compartment and, if so, whether capture can be accomplished from one dendritic compartment to the other. We found that capture of L-LTP can also occur within the basilar dendritic compartment and that the tagging signal that enables capture appears to be the same in both dendritic compartments. However, capture across compartments, between the apical and basilar dendrites, follows different rules and requires a stronger triggering stimulation than capture within a compartment. These results suggest that the tag appears specific to a compartment either apical or basilar and that an additional mechanism may be required to capture across compartments.


Subject(s)
Dendrites/physiology , Long-Term Potentiation/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , Electric Stimulation/methods , In Vitro Techniques , Mice , Mice, Inbred C57BL
11.
Neuron ; 48(1): 123-37, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16202713

ABSTRACT

Expression of VP16-CREB, a constitutively active form of CREB, in hippocampal neurons of the CA1 region lowers the threshold for eliciting the late, persistent phase of long-term potentiation (L-LTP) in the Schaffer collateral pathway. This VP16-CREB-mediated L-LTP differs from the conventional late phase of LTP in not being dependent on new transcription. This finding suggests that in the transgenic mice the mRNA transcript(s) encoding the protein(s) necessary for this form of L-LTP might already be present in CA1 neurons in the basal condition. We used high-density oligonucleotide arrays to identify the mRNAs differentially expressed in the hippocampus of transgenic and wild-type mice. We then explored the contribution of the most prominent candidate genes revealed by our screening, namely prodynorphin, BDNF, and MHC class I molecules, to the facilitated LTP of VP16-CREB mice. We found that the overexpression of brain-derived neurotrophic factor accounts for an important component of this phenotype.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , CREB-Binding Protein/physiology , Herpes Simplex Virus Protein Vmw65/physiology , Long-Term Potentiation/genetics , Neuronal Plasticity/genetics , Synapses/genetics , Animals , Brain-Derived Neurotrophic Factor/deficiency , CREB-Binding Protein/deficiency , Excitatory Postsynaptic Potentials/genetics , Exons , Female , Gene Expression Profiling/methods , Herpes Simplex Virus Protein Vmw65/deficiency , Hippocampus/cytology , In Situ Hybridization/methods , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/physiology , Patch-Clamp Techniques/methods , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Synaptic Transmission , Time Factors
12.
Learn Mem ; 11(3): 318-27, 2004.
Article in English | MEDLINE | ID: mdl-15169862

ABSTRACT

CPEB-1 is a sequence-specific RNA binding protein that stimulates the polyadenylation-induced translation of mRNAs containing the cytoplasmic polyadenylation element (CPE). Although CPEB-1 was identified originally in Xenopus oocytes, it has also been found at postsynaptic sites of hippocampal neurons where, in response to N-methyl-D-aspartate receptor activation, it is thought to induce the polyadenylation and translation of alphaCaMKII and perhaps other CPE-containing mRNAs. Because some forms of synaptic modification appear to be influenced by local (synaptic) protein synthesis, we examined long-term potentiation (LTP) in CPEB-1 knockout mice. Although the basal synaptic transmission of Schaffer collateral-CA1 neurons was not affected in the knockout mice, we found that there was a modest deficit in LTP evoked by a single train of 100 Hz stimulation, but a greater deficit in LTP evoked by one train of theta-burst stimulation. In contrast, LTP evoked by either four trains of 100 Hz stimulation or five trains of theta-burst stimulation were not or were only modestly affected, respectively. The deficit in LTP evoked by single stimulation in knockout mice appeared several minutes after tetanic stimulation. Long-term depression (LTD) evoked by 1 Hz stimulation was moderately facilitated; however, a stronger and more enduring form of LTD induced by paired-pulse 1 Hz stimulation was unaffected. These data suggest that CPEB-1 contributes in the translational control of mRNAs that is critical only for some selected forms of LTP and LTD.


Subject(s)
Hippocampus/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Synapses/metabolism , Transcription Factors/physiology , Xenopus Proteins , mRNA Cleavage and Polyadenylation Factors/physiology , Action Potentials/drug effects , Action Potentials/genetics , Action Potentials/physiology , Animals , Antimetabolites/pharmacology , Electric Stimulation , Hippocampus/cytology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Organ Culture Techniques , RNA/analysis , RNA, Messenger/analysis , Sirolimus/pharmacology , Synapses/drug effects , Synapses/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Transcription Factors/drug effects , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/drug effects , mRNA Cleavage and Polyadenylation Factors/genetics
13.
Neuron ; 42(6): 947-59, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15207239

ABSTRACT

We studied a mouse model of the haploinsufficiency form of Rubinstein-Taybi syndrome (RTS), an inheritable disorder caused by mutations in the gene encoding the CREB binding protein (CBP) and characterized by mental retardation and skeletal abnormalities. In these mice, chromatin acetylation, some forms of long-term memory, and the late phase of hippocampal long-term potentiation (L-LTP) were impaired. We ameliorated the L-LTP deficit in two ways: (1) by enhancing the expression of CREB-dependent genes, and (2) by inhibiting histone deacetyltransferase activity (HDAC), the molecular counterpart of the histone acetylation function of CBP. Inhibition of HDAC also reversed the memory defect observed in fear conditioning. These findings suggest that some of the cognitive and physiological deficits observed on RTS are not simply due to the reduction of CBP during development but may also result from the continued requirement throughout life for both the CREB co-activation and the histone acetylation function of CBP.


Subject(s)
Chromatin/metabolism , Long-Term Potentiation/physiology , Memory/physiology , Nuclear Proteins/metabolism , Rubinstein-Taybi Syndrome/physiopathology , Trans-Activators/metabolism , Acetylation , Analysis of Variance , Animals , Blotting, Western/methods , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein , Cell Line , Chromatin/classification , Conditioning, Psychological , Disease Models, Animal , Dynorphins/metabolism , Electrophysiology , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Fear , Female , Gene Expression , Heterozygote , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Kidney , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Neural Inhibition , Neurons/drug effects , Neurons/physiology , Nuclear Proteins/genetics , Phosphodiesterase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Psychomotor Performance , Reaction Time , Recognition, Psychology , Rolipram/pharmacology , Rubinstein-Taybi Syndrome/genetics , Synaptophysin/metabolism , Time Factors , Trans-Activators/genetics , Transfection
14.
Cell ; 108(5): 689-703, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11893339

ABSTRACT

Restricted and regulated expression in mice of VP16-CREB, a constitutively active form of CREB, in hippocampal CA1 neurons lowers the threshold for eliciting a persistent late phase of long-term potentiation (L-LTP) in the Schaffer collateral pathway. This L-LTP has unusual properties in that its induction is not dependent on transcription. Pharmacological and two-pathway experiments suggest a model in which VP16-CREB activates the transcription of CRE-driven genes and leads to a cell-wide distribution of proteins that prime the synapses for subsequent synapse-specific capture of L-LTP by a weak stimulus. Our analysis indicates that synaptic capture of CRE-driven gene products may be sufficient for consolidation of LTP and provides insight into the molecular mechanisms of synaptic tagging and synapse-specific potentiation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Synapses/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Doxycycline/pharmacology , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Genes, Reporter , Hippocampus/cytology , Hippocampus/metabolism , In Situ Hybridization , In Vitro Techniques , Memory/physiology , Mice , Mice, Transgenic , Models, Biological , Neurons/cytology , Protein Biosynthesis , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...