Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
BMC Biol ; 15(1): 112, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29183317

ABSTRACT

BACKGROUND: Smad4 is a critical effector of TGF-ß signaling that regulates a variety of cellular functions. However, its role in the brain has rarely been studied. Here, we examined the molecular mechanisms underlying the post-translational regulation of Smad4 function by SUMOylation, and its role in spatial memory formation. RESULTS: In the hippocampus, Smad4 is SUMOylated by the E3 ligase PIAS1 at Lys-113 and Lys-159. Both spatial training and NMDA injection enhanced Smad4 SUMOylation. Inhibition of Smad4 SUMOylation impaired spatial learning and memory in rats by downregulating TPM2, a gene associated with skeletal myopathies. Similarly, knockdown of TPM2 expression impaired spatial learning and memory, while TPM2 mRNA and protein expression increased after spatial training. Among the TPM2 mutations associated with skeletal myopathies, the TPM2E122K mutation was found to reduce TPM2 expression and impair spatial learning and memory in rats. CONCLUSIONS: We have identified a novel role of Smad4 SUMOylation and TPM2 in learning and memory formation. These results suggest that patients with skeletal myopathies who carry the TPM2E122K mutation may also have deficits in learning and memory functions.


Subject(s)
Smad4 Protein/genetics , Spatial Memory , Sumoylation , Tropomyosin/genetics , Up-Regulation , Animals , Male , Rats , Rats, Sprague-Dawley , Smad4 Protein/metabolism , Tropomyosin/metabolism
2.
Nat Commun ; 7: 10552, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26842955

ABSTRACT

The methyl-CpG-binding protein 2 (MeCP2) gene, MECP2, is an X-linked gene encoding the MeCP2 protein, and mutations of MECP2 cause Rett syndrome (RTT). However, the molecular mechanism of MECP2-mutation-caused RTT is less known. Here we find that MeCP2 could be SUMO-modified by the E3 ligase PIAS1 at Lys-412. MeCP2 phosphorylation (at Ser-421 and Thr-308) facilitates MeCP2 SUMOylation, and MeCP2 SUMOylation is induced by NMDA, IGF-1 and CRF in the rat brain. MeCP2 SUMOylation releases CREB from the repressor complex and enhances Bdnf mRNA expression. Several MECP2 mutations identified in RTT patients show decreased MeCP2 SUMOylation. Re-expression of wild-type MeCP2 or SUMO-modified MeCP2 in Mecp2-null neurons rescues the deficits of social interaction, fear memory and LTP observed in Mecp2 conditional knockout (cKO) mice. These results together reveal an important role of MeCP2 SUMOylation in social interaction, memory and synaptic plasticity, and that abnormal MeCP2 SUMOylation is implicated in RTT.


Subject(s)
Behavior, Animal , Hippocampus/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , Rett Syndrome/metabolism , Sumoylation , Animals , Blotting, Western , CA1 Region, Hippocampal/metabolism , Disease Models, Animal , Exploratory Behavior , Fear , Gene Knockout Techniques , Genetic Vectors , Immunohistochemistry , Immunoprecipitation , In Vitro Techniques , Learning , Lentivirus , Memory , Methyl-CpG-Binding Protein 2/genetics , Mice , Mutagenesis, Site-Directed , Mutation , Neuronal Plasticity , Protein Inhibitors of Activated STAT/metabolism , Rats , Rats, Sprague-Dawley , Rett Syndrome/genetics , Reverse Transcriptase Polymerase Chain Reaction , Social Behavior
3.
Neuropsychopharmacology ; 36(12): 2571-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21849984

ABSTRACT

Laminin is a major structural element of the basal lamina consisting of an α-chain, a ß-chain, and a γ-chain arranged in a cross-like structure, with their C-terminal inter-coiled. Laminin is abundantly expressed in the hippocampus of mature brain and is implicated in several psychiatric disorders, but its possible role involved in learning and memory function is not known. This issue was examined here. Our results revealed that water maze training significantly decreased laminin-ß1 (LB1) expression in the rat hippocampal CA1 area. Transfection of LB1 WT plasmid to hippocampal CA1 neurons impaired water maze performance in rats. Meanwhile, it decreased the phosphorylation level of ERK/MAPK and protein kinase serum- and glucocorticoid-inducible kinase-1 (SGK1). By contrast, knockdown of endogenous LB1 expression using RNA interference (LB1 siRNA) enhanced water maze performance. Meanwhile, it increased the phosphorylation level of ERK/MAPK and SGK1. The enhancing effect of LB1 siRNA on spatial learning and on the phosphorylation of ERK/MAPK and SGK1 was blocked by co-treatment with the MEK inhibitor U0126 at a concentration that did not apparently affect spatial learning and ERK/MAPK phosphorylation alone. Further, the enhancing effect of LB1 siRNA on spatial learning and SGK1 phosphorylation was similarly blocked by co-transfection with SGK1 siRNA at a concentration that did not markedly affect spatial learning and SGK1 expression alone. These results together indicate that LB1 negatively regulates spatial learning in rats. In addition, LB1 impairs spatial learning through decreased activation of the ERK/MAPK-SGK1 signaling pathway in the rat hippocampus.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Immediate-Early Proteins/antagonists & inhibitors , Laminin/physiology , MAP Kinase Signaling System/physiology , Maze Learning/physiology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Knockdown Techniques , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Male , Mitogen-Activated Protein Kinases/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Rats , Rats, Sprague-Dawley
4.
Brain Pathol ; 21(2): 150-62, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20731656

ABSTRACT

Brain-derived neurotrophic factor (BDNF) was shown to produce its neuroprotective effect through extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphatidylinositol-3 kinase (PI3-K) signaling. But whether other pathways also mediate the neuroprotective effect of BDNF is less known. In this study, we found that direct administration of BDNF to rat hippocampal CA1 area dose-dependently increased the mRNA and protein levels of Bcl-xL. BDNF also increased protein kinase casein kinase II (CK2) activity and NF-κB phosphorylation at Ser529 dose-dependently. Further, transfection of the wild-type CK2α DNA to CA1 neurons increased nuclear factor kappa B (NF-κB) phosphorylation and Bcl-xL mRNA expression, whereas transfection of CK2α156A, the catalytically inactive mutant of CK2α, decreased these measures. Moreover, transfection of CK2α small interfering RNA (siRNA) blocked the enhancing effect of BDNF on NF-κB phosphorylation and Bcl-xL expression. These results were further confirmed by treatment of 4,5,6,7-tetrabromobenzotriazole (TBB), a specific CK2 inhibitor. Transfection of NF-κBS529A, the dominant negative mutant of NF-κB, prevented the enhancing effect of BDNF on Bcl-xL expression. More importantly, BDNF activation of CK2 is not affected by co-administration of the ERK1/2 inhibitor, PD98059, and the PI3-K inhibitor, LY294002. These results demonstrate a novel BDNF signaling pathway and provide an alternative therapeutic strategy for the protective effect of BDNF on hippocampal neurons in vivo.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Casein Kinase II/metabolism , Hippocampus/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , bcl-X Protein/metabolism , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/pharmacology , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Immunohistochemistry , In Situ Nick-End Labeling , Male , RNA Interference , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
EMBO J ; 30(1): 205-20, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21102409

ABSTRACT

By using differential display PCR, we have previously identified 98 cDNA fragments from rat dorsal hippocampus, which are expressed differentially between the fast learners and slow learners from water-maze learning task. One cDNA fragment, which showed a higher expression level in fast learners, encodes the rat protein inhibitor of activated STAT1 (pias1) gene. Spatial training induced a significant increase in PIAS1 expression in rat hippocampus. Transient transfection of the wild-type (WT) PIAS1 plasmid to CA1 neurons facilitated, whereas transfection of PIAS1 siRNA impaired spatial learning in rats. Meanwhile, PIAS1WT increased STAT1 sumoylation, decreased STAT1 DNA binding and decreased STAT1 phosphorylation at Tyr-701 associated with spatial learning facilitation. But PIAS1 siRNA transfection produced an opposite effect on these measures associated with spatial learning impairment. Further, transfection of STAT1 sumoylation mutant impaired spatial acquisition, whereas transfection of STAT1 phosphorylation mutant blocked the impairing effect of PIAS1 siRNA on spatial learning. In this study, we first demonstrate the role of PIAS1 in spatial learning. Both posttranslational modifications (increased sumoylation and decreased phosphorylation) mediate the effect of PIAS1 on spatial learning facilitation.


Subject(s)
Maze Learning , Protein Inhibitors of Activated STAT/metabolism , STAT1 Transcription Factor/metabolism , Animals , Cells, Cultured , DNA/metabolism , Gene Expression Regulation , Hippocampus/cytology , Hippocampus/metabolism , Male , Mutation , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/genetics , Sumoylation , Transfection
6.
J Neurosci ; 27(23): 6243-8, 2007 Jun 06.
Article in English | MEDLINE | ID: mdl-17553997

ABSTRACT

Casein kinase II (CK2) is a multifunctional serine/threonine protein kinase that is associated with the development of neuritogenesis and synaptic plasticity. The phosphoinositide 3-kinase (PI-3K)/Akt pathway is implicated in long-term memory formation. In addition, serum- and glucocorticoid-inducible kinase 1 (SGK1) is another downstream target of PI-3K signaling that was shown to play an important role in spatial memory formation. Whether CK2 may also affect memory formation and whether CK2 interacts with Akt and SGK1 during this process is unknown. In the present study, we found that water maze training significantly decreased CK2 activity in the rat hippocampal CA1 area but not in the dentate gyrus (DG) area. Transfection of the dominant negative mutant of CK2, CK2alphaA(156), to the CA1 area, but not to the DG area, decreased CK2 activity but enhanced spatial memory formation. Meanwhile, it increased SGK1 phosphorylation at Ser422, decreased Akt phosphorylation at Ser473, and increased cAMP response element-binding protein phosphorylation at Ser133. Transfection of the constitutively active SGK1, SGKS422D, enhanced whereas transfection of the wild-type Akt impaired spatial memory formation. Also, administration of the protein phosphatase 2A inhibitor, fostriecin, reversed the memory-impairing effect of CK2alphaWT. It also reversed the effect of CK2alphaWT in decreasing SGK1 phosphorylation. Akt Ser473 phosphorylation was moderately increased by CK2alphaWT and fostriecin treatment, but AktS473A mutant transfection reversed the memory-impairing effect of CK2alphaWT. These results together suggest that CK2 impairs spatial memory formation through differential cross talk with PI-3 kinase signaling by activation of Akt and inactivation of SGK1 through protein phosphatase 2A.


Subject(s)
Casein Kinase II/metabolism , Down-Regulation , Immediate-Early Proteins/antagonists & inhibitors , Maze Learning/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Casein Kinase II/genetics , Down-Regulation/genetics , Enzyme Activation/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Receptor Cross-Talk/physiology , Signal Transduction/genetics , Spatial Behavior/physiology
7.
J Neurochem ; 100(6): 1531-42, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17241237

ABSTRACT

Mitogen-activated protein kinase/extracellular signal-regulated kinase plays an important role in memory formation and directly phosphorylates serum- and glucocorticoid-inducible kinase1 (SGK1) at Ser78. In this study, we examined the role and mechanism of SGK1 Ser78 activation involved in contextual fear memory formation in rats. Results revealed that SGK1 Ser78 phosphorylation was increased 30 min, 1 h and 3 h after training. Transient transfection of the dominant negative mutant of sgk, sgkS78A, to hippocampal neurons impaired, whereas transfection of the constitutively active sgk, sgkS78D, enhanced fear retention. Microarray analysis identified 14 genes that showed more than threefold alteration in their gene expression in sgkS78A-transfected animals 6 h after training. One of them is Hairy and Enhancer of split 5 (Hes5). The expression level of Hes5 is approximately 4.4-fold higher in sgkS78A-transfected animals. Further analyses revealed that Hes5 level is markedly decreased after training in control animals, but sgkS78A markedly increased Hes5 level after training. RNA interference experiment showed that shHes5 dose-dependently enhanced fear retention, whereas over-expression of Hes5 impaired fear retention. Moreover, shHes5 at a lower concentration completely blocked the memory-impairing effect of sgkS78A. These results together suggest that Hes5 negatively regulates contextual fear memory formation and SGK activation down-regulates Hes5 expression to enhance fear retention.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation/physiology , Fear , Immediate-Early Proteins/metabolism , Memory/physiology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Acoustic Stimulation/adverse effects , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Behavior, Animal/drug effects , Conditioning, Classical/physiology , Gene Transfer Techniques , Green Fluorescent Proteins/metabolism , Immediate-Early Proteins/genetics , Male , Motor Activity/genetics , PC12 Cells , Protein Serine-Threonine Kinases/genetics , RNA Interference/physiology , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Serine/genetics , Time Factors
8.
Learn Mem ; 13(2): 114-8, 2006.
Article in English | MEDLINE | ID: mdl-16585788

ABSTRACT

Previous studies showed that the serum- and glucocorticoid-inducible kinase (sgk) gene plays an important role in long-term memory formation. The present study further examined the role of SGK in long-term potentiation (LTP). The dominant-negative mutant of sgk, SGKS422A, was used to inactivate SGK. Results revealed a time-dependent increase in SGK phosphorylation after tetanization with a significant effect observed 3 h and 5 h later. Transfection of SGKS422A impaired the expression, but not the induction, of LTP. Furthermore, the constitutively active sgk, SGKS422D, up-regulated postsynaptic density-95 expression in the hippocampus. These results together support the role of SGK in neuronal plasticity.


Subject(s)
Hippocampus/physiology , Immediate-Early Proteins/physiology , Long-Term Potentiation/physiology , Neurons/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Hippocampus/cytology , Neuronal Plasticity/physiology , Phosphorylation , Rats , Rats, Mutant Strains , Synaptic Transmission/physiology , Time Factors , Transfection
9.
Eur J Neurosci ; 23(5): 1311-20, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16553792

ABSTRACT

We have previously demonstrated that serum- and glucocorticoid-inducible kinase (SGK) plays a causal role in facilitating memory formation of spatial learning in rats, but the SGK signaling pathway involved in spatial memory formation is not known. The mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) also plays an important role in memory formation. We therefore examined whether SGK is a downstream target of the MAPK/ERK signaling cascade and whether ERK signaling to SGK mediates spatial memory formation in rats. Results from an in vitro kinase assay revealed that ERK directly phosphorylates SGK at Ser78, but not at Thr256 and Ser422, whereas inhibition of ERK by PD98059 significantly decreased SGK phosphorylation at Ser78, Thr256 and Ser422 following spatial training. Prior administration of PD98059 also antagonized the enhancing effect of 12-O-tetradecanoylphorbol-13-acetate (TPA), a protein kinase C activator that also causes ERK activation, on SGK phosphorylation and cAMP response element binding protein (CREB) phosphorylation. Moreover, TPA-induced SGK phosphorylation and CREB phosphorylation was abolished by prior SGKS78A mutant DNA transfection. By contrast, SGKS78A mutant DNA transfection to hippocampal area CA1 did not affect spatial memory formation, whereas SGKT256A mutant DNA transfection to area CA1 significantly impaired spatial memory formation. ERK was known to regulate sgk mRNA expression, but in the present study we have demonstrated that SGK is also a downstream target of the ERK signaling cascade; ERK directly phosphorylates SGK at Ser78 and indirectly activates SGK at Thr256 and Ser422 through unknown intermediate molecules. Furthermore, ERK activation of SGK is involved in spatial memory formation in rats.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Immediate-Early Proteins/metabolism , MAP Kinase Signaling System/physiology , Memory/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/metabolism , Immediate-Early Proteins/genetics , Male , Maze Learning/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/metabolism
10.
Neurobiol Aging ; 27(1): 105-18, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16298246

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is suggested as a specific neurotrophic factor for midbrain dopamine (DA) neurons, but the molecular mechanism underlying the neuroprotective action of GDNF is not well known. In the present study, we have shown that GDNF increased protein kinase CK2 activity in rat substantia nigra (SN) in a dose-dependent and time-dependent manner. This effect is prevented by prior treatment of the receptor Ret blocker K-252b. Immunostaining results also revealed that CK2 is expressed in TH-positive neurons in mesencephalon culture. Transfection of the wildtype CK2alpha DNA increased, whereas transfection of the catalytically inactive CK2alphaA156 mutant DNA decreased CK2 activity in the SN. CK2alphaA156 mutant DNA also antagonized the enhancing effect of GDNF on CK2 activity. It also antagonized the enhancing effects of GDNF on tyrosine hydroxylase (TH) protein level in the SN, DA turnover in the striatum and rotarod performance in rats. Further, CK2alpha wildtype DNA increased, whereas CK2alphaA156 mutant DNA decreased TH activity in the SN without altering the TH protein level. On the other hand, the DA neuron toxin 1-methyl-4-phenylpyridinium iodide (MPP+) markedly decreased the number of TH-positive neurons and TH protein level in the SN, decreased DA level in the striatum and impaired rotarod performance in rats. Over-expression of the CK2alpha wildtype DNA partially, but significantly, prevented the deteriorating effect of MPP+ on these measures. Prior administration of MPP+ also antagonized the enhancing effect of GDNF on CK2 activity. These results together suggest that the CK2 signaling pathway contributes to the neuroprotective action of GDNF on DA neurons.


Subject(s)
Casein Kinase II/metabolism , Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Motor Activity , Neurons/metabolism , Signal Transduction/physiology , Substantia Nigra/physiopathology , Animals , Dose-Response Relationship, Drug , Male , Nerve Growth Factors/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley , Rotarod Performance Test , Signal Transduction/drug effects , Substantia Nigra/drug effects
11.
J Neurosci ; 23(10): 4072-80, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12764094

ABSTRACT

Tyrosine kinase phosphorylation plays an important role in the induction of long-term potentiation (LTP). Focal adhesion kinase (FAK) is a 125 kDa nonreceptor tyrosine kinase that shows decreased phosphorylation in fyn mutant mice, and Fyn plays a critical role in LTP induction. By examining the role of FAK involved in LTP induction in dentate gyrus in vivo with medial perforant path stimulation, we found that both FAK and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) phosphorylation were increased significantly 5 and 10 min after LTP induction, whereas cAMP-responsive element binding protein (CREB) phosphorylation was increased 40 min later. Transfection of the dominant-negative FAK mutant construct HA-FAK(Y397F) impaired LTP, whereas transfection of the constitutively activated form HA-FAK(Delta1-100) reduced the threshold for LTP induction. Transfection of HA-FAK(Delta1-100) by itself did not induce long-lasting potentiation. Further, transfection of the HA-FAK(Y397F) construct decreased FAK, MAPK/ERK, and CREB phosphorylation, and the inhibition of MAPK/ERK decreased CREB phosphorylation. Moreover, blockade of NMDA receptor (NMDAR) did not decrease FAK, MAPK/ERK, and CREB phosphorylation although LTP induction was blunted by NMDAR antagonist. These biochemical changes were not associated with low-frequency stimulation either. Immunoprecipitation results revealed that tyrosine phosphorylation of NR2A and NR2B as well as the association of phosphorylated FAK with NR2A and NR2B was increased with LTP induction. These results together suggest that FAK is required, but not sufficient, for the induction of LTP in a NMDAR-independent manner and that MAPK/ERK and CREB are the downstream events of FAK activation. Further, FAK may interact with NR2A and NR2B to modulate LTP induction.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/enzymology , Long-Term Potentiation/physiology , Neurons/enzymology , Neurons/physiology , Protein-Tyrosine Kinases/physiology , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/physiology , Dentate Gyrus/chemistry , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Mutagenesis, Site-Directed , Neurons/chemistry , Phosphorylation , Plasmids , Precipitin Tests , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-fyn , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Reaction Time/genetics , Reaction Time/physiology , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Sensory Thresholds/physiology , Synaptosomes/metabolism , Transfection
12.
Proc Natl Acad Sci U S A ; 99(6): 3990-5, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11891330

ABSTRACT

By using differential display PCR, we have identified 98 cDNA fragments from the rat dorsal hippocampus that are expressed differentially between the fast learners and slow learners in the water maze learning task. One of these cDNA fragments encodes the rat serum- and glucocorticoid-inducible kinase (sgk) gene. Northern blot analysis revealed that the sgk mRNA level was approximately 4-fold higher in the hippocampus of fast learners than slow learners. In situ hybridization results indicated that sgk mRNA level was increased markedly in CA1, CA3, and dentate gyrus of hippocampus in fast learners. Transient transfection of the sgk mutant DNA to the CA1 area impaired, whereas transfection of the sgk wild-type DNA facilitated water maze performance in rats. These results provide direct evidence that enhanced sgk expression facilitates memory consolidation of spatial learning in rats. These results also elucidate the molecular mechanism of glucocorticoid-induced memory facilitation in mammals.


Subject(s)
Glucocorticoids/pharmacology , Hippocampus/metabolism , Memory/physiology , Nuclear Proteins , Protein Serine-Threonine Kinases/metabolism , Space Perception/physiology , Animals , Base Sequence , Blotting, Northern , Enzyme Induction/drug effects , Gene Expression Profiling , Immediate-Early Proteins , In Situ Hybridization , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Molecular Sequence Data , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Space Perception/drug effects , Time Factors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...