Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 273, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900294

ABSTRACT

Long-term memory formation requires de novo RNA and protein synthesis. Using differential display PCR, we found that the NCoR1 cDNA fragment is differentially expressed between fast learners and slow learners, with fast learners showing a lower expression level than slow learners in the water maze learning task. Fast learners also show lower NCoR1 mRNA and protein expression levels. In addition, spatial training decreases both NCoR1 mRNA and protein expression, whereas NCoR1 conditional knockout (cKO) mice show enhanced spatial memory. In studying the molecular mechanism, we found that spatial training decreases the association between NCoR1 and DEC2. Both NCoR1 and DEC2 suppress the expression of BDNF, integrin α3 and SGK1 through C/EBPα binding to their DNA promoters, but overexpression of DEC2 in NCoR1 cKO mice rescues the decreased expression of these proteins compared with NCoR1 loxP mice overexpressing DEC2. Further, spatial training decreases DEC2 expression. Spatial training also enhances C/EBPα binding to Bdnf, Itga3 and Sgk1 promoters, an effect also observed in fast learners, and both NCoR1 and DEC2 control C/EBPα activity. Whereas knockdown of BDNF, integrin α3 or SGK1 expression impairs spatial learning and memory, it does not affect Y-maze performance, suggesting that BDNF, integrin α3 and SGK1 are involved in long-term memory formation, but not short-term memory formation. Moreover, NCoR1 expression is regulated by the JNK/c-Jun signaling pathway. Collectively, our findings identify DEC2 as a novel interacting protein of NCoR1 and elucidate the novel roles and mechanisms of NCoR1 and DEC2 in negative regulation of spatial memory formation.


Subject(s)
Maze Learning , Mice, Knockout , Nuclear Receptor Co-Repressor 1 , Spatial Memory , Animals , Spatial Memory/physiology , Mice , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Maze Learning/physiology , Male , Mice, Inbred C57BL , Promoter Regions, Genetic , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Protein Serine-Threonine Kinases , Immediate-Early Proteins
2.
PLoS One ; 18(4): e0283908, 2023.
Article in English | MEDLINE | ID: mdl-37023120

ABSTRACT

Long-term memory formation requires de novo RNA and protein synthesis. By using the differential display-polymerase chain reaction strategy, we have presently identified the Nedd4 family interacting protein 1 (Ndfip1) cDNA fragment that is differentially expressed between the slow learners and the fast learners from the water maze learning task in rats. Further, the fast learners show decreased Ndfip1 mRNA and protein expression levels than the slow learners. Spatial training similarly decreases the Ndfip1 mRNA and protein expression levels. Conversely, the Ndfip1 conditional heterozygous (cHet) mice show enhanced spatial memory performance compared to the Ndfip1flox/WT control mice. Result from co-immunoprecipitation experiment indicates that spatial training decreases the association between Ndfip1 and the E3 ubiquitin ligase Nedd4 (Nedd4-1), and we have shown that both Beclin 1 and PTEN are endogenous ubiquitination targets of Nedd4 in the hippocampus. Further, spatial training decreases endogenous Beclin 1 and PTEN ubiquitination, and increases Beclin 1 and PTEN expression in the hippocampus. On the other hand, the Becn1 conditional knockout (cKO) mice and the Pten cKO mice both show impaired spatial learning and memory performance. Moreover, the expression level of Beclin 1 and PTEN is higher in the Ndfip1 cHet mice compared with the Ndfip1flox/WT control mice. Here, we have identified Ndfip1 as a candidate novel negative regulation for spatial memory formation and this is associated with increased ubiquitination of Beclin 1 and PTEN in the hippocampus.


Subject(s)
Carrier Proteins , Endosomal Sorting Complexes Required for Transport , Animals , Mice , Rats , Beclin-1/metabolism , Carrier Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Nedd4 Ubiquitin Protein Ligases/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Messenger/metabolism , Spatial Memory , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
3.
Mol Ther ; 29(1): 376-395, 2021 01 06.
Article in English | MEDLINE | ID: mdl-32950104

ABSTRACT

The amyloid precursor protein (APP) intracellular domain (AICD) is implicated in the pathogenesis of Alzheimer's disease (AD), but post-translational modification of AICD has rarely been studied and its role in AD is unknown. In this study, we examined the role and molecular mechanism of AICD SUMOylation in the pathogenesis of AD. We found that AICD is SUMO-modified by the SUMO E3 ligase protein inhibitor of activated STAT1 (PIAS1) in the hippocampus at Lys-43 predominantly, and that knockdown of PIAS1 decreases endogenous AICD SUMOylation. AICD SUMOylation increases AICD association with its binding protein Fe65 and increases AICD nuclear translocation. Furthermore, AICD SUMOylation increases AICD association with cyclic AMP-responsive element binding protein (CREB) and p65 and their DNA binding for transcriptional activation of neprilysin (NEP) and transthyretin (TTR), two major Aß-degrading enzymes, respectively. Consequently, AICD SUMOylation decreases the Aß level, Aß oligomerization, and amyloid plaque deposits. It also rescues spatial memory deficits in APP/PS1 mice. Conversely, blockade of AICD SUMOylation at Lys-43 produces the opposite effects. Melatonin is identified as an endogenous stimulus that induces AICD SUMOylation. It also decreases the Aß level and rescues reduction of PIAS1, NEP, and TTR expression in APP/PS1 mice. In this study, we demonstrate that AICD SUMOylation functions as a novel endogenous defense mechanism to combat AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Melatonin/metabolism , Transcriptional Activation , Alzheimer Disease/pathology , Animals , CREB-Binding Protein/metabolism , Disease Models, Animal , Melatonin/pharmacology , Mice , Protein Binding , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , Proteolysis/drug effects , Sumoylation/drug effects , Transcriptional Activation/drug effects
4.
Sci Rep ; 10(1): 1074, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31974426

ABSTRACT

The methyl-CpG-binding protein 2 gene, MECP2, is an X chromosome-linked gene encoding the MeCP2 protein, and mutations of MECP2 cause Rett syndrome (RTT). Previous study has shown that re-expression of SUMO-modified MeCP2 in Mecp2-null neurons rescues synaptic and behavioral deficits in Mecp2 conditional knockout mice, whereas about 12-fold decrease in Wnt6 mRNA level was found in MeCP2K412R sumo-mutant mice. Here, we examined the role of Wnt6 in MeCP2 T158A mouse model of RTT. Results show that lentiviral delivery of Wnt6 to the amygdala ameliorates locomotor impairment and social behavioral deficits in these animals. MeCP2 T158A mice show decreased level of GSK-3ß phosphorylation and increased level of ß-catenin phosphorylation. They also show reduced level of MeCP2 SUMOylation. These alterations were also restored by lenti-Wnt6 transduction. Further, both BDNF and IGF-1 expressions are decreased in MeCP2 T158A mice. Overexpression of Wnt6 increases Bdnf and Igf-1 promoter activity in HEK293T cells in a dose-dependent manner. Lenti-Wnt6 transduction to the amygdala similarly increases the mRNA level and protein expression of BDNF and IGF-1 in MeCP2 T158A mice. Moreover, environmental enrichment (EE) similarly ameliorates the locomotor and social behavioral deficits in MeCP2 T158A mice. One of the mechanisms underlying EE is mediated through enhanced MeCP2 SUMOylation and increased Wnt6 expression in these animals by EE.


Subject(s)
Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Proto-Oncogene Proteins/metabolism , Rett Syndrome/metabolism , Wnt Proteins/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Proto-Oncogene Proteins/genetics , Rett Syndrome/genetics , Signal Transduction , Wnt Proteins/genetics
5.
Cell Death Differ ; 27(1): 192-209, 2020 01.
Article in English | MEDLINE | ID: mdl-31127200

ABSTRACT

Amyloid-ß (Aß) oligomers largely initiate the cascade underlying the pathology of Alzheimer's disease (AD). Galectin-3 (Gal-3), which is a member of the galectin protein family, promotes inflammatory responses and enhances the homotypic aggregation of cancer cells. Here, we examined the role and action mechanism of Gal-3 in Aß oligomerization and Aß toxicities. Wild-type (WT) and Gal-3-knockout (KO) mice, APP/PS1;WT mice, APP/PS1;Gal-3+/- mice and brain tissues from normal subjects and AD patients were used. We found that Aß oligomerization is reduced in Gal-3 KO mice injected with Aß, whereas overexpression of Gal-3 enhances Aß oligomerization in the hippocampi of Aß-injected mice. Gal-3 expression shows an age-dependent increase that parallels endogenous Aß oligomerization in APP/PS1 mice. Moreover, Aß oligomerization, Iba1 expression, GFAP expression and amyloid plaque accumulation are reduced in APP/PS1;Gal-3+/- mice compared with APP/PS1;WT mice. APP/PS1;Gal-3+/- mice also show better acquisition and retention performance compared to APP/PS1;WT mice. In studying the mechanism underlying Gal-3-promoted Aß oligomerization, we found that Gal-3 primarily co-localizes with Iba1, and that microglia-secreted Gal-3 directly interacts with Aß. Gal-3 also interacts with triggering receptor expressed on myeloid cells-2, which then mediates the ability of Gal-3 to activate microglia for further Gal-3 expression. Immunohistochemical analyses show that the distribution of Gal-3 overlaps with that of endogenous Aß in APP/PS1 mice and partially overlaps with that of amyloid plaque. Moreover, the expression of the Aß-degrading enzyme, neprilysin, is increased in Gal-3 KO mice and this is associated with enhanced integrin-mediated signaling. Consistently, Gal-3 expression is also increased in the frontal lobe of AD patients, in parallel with Aß oligomerization. Because Gal-3 expression is dramatically increased as early as 3 months of age in APP/PS1 mice and anti-Aß oligomerization is believed to protect against Aß toxicity, Gal-3 could be considered a novel therapeutic target in efforts to combat AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloidogenic Proteins/metabolism , Galectin 3/physiology , Age Factors , Alzheimer Disease/psychology , Amyloid beta-Peptides , Animals , Blood Proteins/metabolism , Calcium-Binding Proteins , Disease Models, Animal , Female , Galectin 3/genetics , Galectin 3/metabolism , Galectins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Integrins/metabolism , Male , Memory , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins , Neprilysin/metabolism , Peptide Fragments , Plaque, Amyloid , Rats, Sprague-Dawley , Signal Transduction
6.
Br J Pharmacol ; 176(11): 1793-1810, 2019 06.
Article in English | MEDLINE | ID: mdl-30849179

ABSTRACT

BACKGROUND AND PURPOSE: Protein inhibitor of activated STAT1 (PIAS1) is phosphorylated by IKKα at Ser90 in a PIAS1 E3 ligase activity-dependent manner. Whether PIAS1 is also phosphorylated at other residues and the functional significance of these additional phosphorylation events are not known. The transcription factor Elk-1 remains SUMOylated under basal conditions, but the role of Elk-1 SUMOylation in brain is unknown. Here, we examined the functional significance of PIAS1-mediated Elk-1 SUMOylation in Alzheimer's disease (AD) using the APP/PS1 mouse model of AD and amyloid ß (Aß) microinjections in vivo. EXPERIMENTAL APPROACH: Novel phosphorylation site(s) on PIAS1 were identified by LC-MS/MS, and MAPK/ERK-mediated phosphorylation of Elk-1 demonstrated using in vitro kinase assays. Elk-1 SUMOylation by PIAS1 in brain was determined using in vitro SUMOylation assays. Apoptosis in hippocampus was assessed by measuring GADD45α expression by western blotting, and apoptosis of hippocampal neurons in APP/PS1 mice was assessed by TUNEL assay. KEY RESULTS: Using LC-MS/MS, we identified a novel MAPK/ERK-mediated phosphorylation site on PIAS1 at Ser503 and showed this phosphorylation determines PIAS1 E3 ligase activity. In rat brain, Elk-1 was SUMOylated by PIAS1, which decreased Elk-1 phosphorylation and down-regulated GADD45α expression. Moreover, lentiviral-mediated transduction of Elk-1-SUMO1 reduced the number of hippocampal apoptotic neurons in APP/PS1 mice. CONCLUSIONS AND IMPLICATIONS: MAPK/ERK-mediated phosphorylation of PIAS1 at Ser503 determines PIAS1 E3 ligase activity. Moreover, PIAS1 mediates SUMOylation of Elk-1, which functions as an endogenous defence mechanism against Aß toxicity in vivo. Targeting Elk-1 SUMOylation could be considered a novel therapeutic strategy against AD.


Subject(s)
Hippocampus/metabolism , Protein Inhibitors of Activated STAT/metabolism , ets-Domain Protein Elk-1/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Phosphorylation , Rats, Sprague-Dawley , Sumoylation , Ubiquitin-Protein Ligases/metabolism
7.
Front Mol Neurosci ; 10: 217, 2017.
Article in English | MEDLINE | ID: mdl-28744198

ABSTRACT

Galectin-3, a member of the galectin protein family, has been found to regulate cell proliferation, inhibit apoptosis and promote inflammatory responses. Galectin-3 is also expressed in the adult rat hippocampus, but its role in learning and memory function is not known. Here, we found that contextual fear-conditioning training, spatial training or injection of NMDA into the rat CA1 area each dramatically decreased the level of endogenous galectin-3 expression. Overexpression of galectin-3 impaired fear memory, whereas galectin-3 knockout (KO) enhanced fear retention, spatial memory and hippocampal long-term potentiation. Galectin-3 was further found to associate with integrin α3, an association that was decreased after fear-conditioning training. Transfection of the rat CA1 area with small interfering RNA against galectin-3 facilitated fear memory and increased phosphorylated focal adhesion kinase (FAK) levels, effects that were blocked by co-transfection of the FAK phosphorylation-defective mutant Flag-FAKY397F. Notably, levels of serine-phosphorylated galectin-3 were decreased by fear conditioning training. In addition, blockade of galectin-3 phosphorylation at Ser-6 facilitated fear memory, whereas constitutive activation of galectin-3 at Ser-6 impaired fear memory. Interestingly galectin-1 plays a role in fear-memory formation similar to that of galectin-3. Collectively, our data provide the first demonstration that galectin-3 is a novel negative regulator of memory formation that exerts its effects through both extracellular and intracellular mechanisms.

8.
Cell Death Differ ; 24(4): 597-614, 2017 04.
Article in English | MEDLINE | ID: mdl-28186506

ABSTRACT

Amyloid-ß (Aß) produces neurotoxicity in the brain and causes neuronal death, but the endogenous defense mechanism that is activated on Aß insult is less well known. Here we found that acute Aß increases the expression of PIAS1 and Mcl-1 via activation of MAPK/ERK, and Aß induction of PIAS1 enhances HDAC1 SUMOylation in rat hippocampus. Knockdown of PIAS1 decreases endogenous HDAC1 SUMOylation and blocks Aß induction of Mcl-1. Sumoylated HDAC1 reduces it association with CREB, increases CREB binding to the Mcl-1 promoter and mediates Aß induction of Mcl-1 expression. Transduction of SUMO-modified lenti-HDAC1 vector to the hippocampus of APP/PS1 mice rescues spatial learning and memory deficit and long-term potentiation impairment in APP/PS1 mice. It also reduces the amount of amyloid plaque and the number of apoptotic cells in CA1 area of APP/PS1 mice. Meanwhile, HDAC1 SUMOylation decreases HDAC1 binding to the neprilysin promoter. These results together reveal an important role of HDAC1 SUMOylation as a naturally occurring defense mechanism protecting against Aß toxicity and provide an alternative therapeutic strategy against AD.


Subject(s)
Amyloid beta-Peptides/toxicity , Epigenesis, Genetic , Histone Deacetylase 1/metabolism , Neuroprotection/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Disease Models, Animal , Gene Expression/drug effects , HEK293 Cells , Hippocampus/metabolism , Histone Deacetylase 1/genetics , Humans , Male , Mice , Mice, Transgenic , Nitriles/pharmacology , Protein Binding , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , RNA Interference , Rats , Rats, Sprague-Dawley , Sumoylation
9.
J Neurosci ; 34(29): 9574-89, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25031400

ABSTRACT

cAMP-responsive element binding protein (CREB) phosphorylation and signaling plays an important role in long-term memory formation, but other posttranslational modifications of CREB are less known. Here, we found that CREB1Δ, the short isoform of CREB, could be sumoylated by the small ubiquitin-like modifier (SUMO) E3 ligase protein inhibitor of activated STAT1 (PIAS1) at Lys271 and Lys290 and PIAS1 SUMOylation of CREB1Δ increased the expression level of CREB1Δ. CREB1Δ could also be sumoylated by other PIAS family proteins, but not by the E3 ligases RanBP2 and Pc2 or by the E2 ligase Ubc9. Furthermore, water maze training increased the level of endogenous CREB SUMOylation in rat CA1 neurons determined by in vitro SUMOylation assay, but this effect was not observed in other brain areas. Moreover, transduction of Lenti-CREBWT to rat CA1 area facilitated, whereas transduction of Lenti-CREB double sumo-mutant (CREBK271RK290R) impaired, spatial learning and memory performance. Transduction of Lenti-CREBWT-SUMO1 fusion vector to rat CA1 area showed a more significant effect in enhancing spatial learning and memory and CREB SUMOylation. Lenti-CREBWT transduction increased, whereas Lenti-CREBK271RK290R transduction decreased, CREB DNA binding to the brain-derived neurotrophic factor (bdnf) promoter and decreased bdnf mRNA expression. Knock-down of PIAS1 expression in CA1 area by PIAS1 siRNA transfection impaired spatial learning and memory and decreased endogenous CREB SUMOylation. In addition, CREB SUMOylation was CREB phosphorylation dependent and lasted longer. Therefore, CREB phosphorylation may be responsible for signal transduction during the early phase of long-term memory formation, whereas CREB SUMOylation sustains long-term memory.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Memory/physiology , Protein Inhibitors of Activated STAT/metabolism , Space Perception/physiology , Sumoylation/physiology , Amino Acid Sequence , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hippocampus/metabolism , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Protein Binding/genetics , Protein Inhibitors of Activated STAT/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Sumoylation/drug effects , Sumoylation/genetics , Time Factors
10.
Neuropsychopharmacology ; 39(3): 746-58, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24081304

ABSTRACT

Signal transducer and activator of transcription-1 (STAT1) has an important role in inflammation and the innate immune response, but its role in the central nervous system is less well understood. Here, we examined the role of STAT1 in spatial learning and memory, and assessed the involvement of STAT1 in mediating the memory-impairing effect of amyloid-beta (Aß). We found that water maze training downregulated STAT1 expression in the rat hippocampal CA1 area, and spatial learning and memory function was enhanced in Stat1-knockout mice. Conversely, overexpression of STAT1 impaired water maze performance. STAT1 strongly upregulated the expression of the extracellular matrix protein laminin ß1 (LB1), which also impaired water maze performance in rats. Furthermore, Aß impaired spatial learning and memory in association with a dose-dependent increase in STAT1 and LB1 expression, but knockdown of STAT1 and LB1 both reversed this effect of Aß. This Aß-induced increase in STAT1 and LB1 expression was also associated with a decrease in the expression of the N-methyl-D-aspartate receptor (NMDAR) subunits, NR1, and NR2B. Overexpression of NR1 or NR2B or exogenous application of NMDA reversed Aß-induced learning and memory deficits as well as Aß-induced STAT1 and LB1 expression. Our results demonstrate that STAT1 negatively regulates spatial learning and memory through transcriptional regulation of LB1 expression. We also identified a novel mechanism for Aß pathogenesis through STAT1 induction. Notably, impairment of spatial learning and memory by this STAT1-mediated mechanism is independent of cAMP responsive element-binding protein signaling.


Subject(s)
Amyloid beta-Peptides/pharmacology , Gene Expression Regulation/drug effects , Memory Disorders/chemically induced , Memory Disorders/metabolism , STAT1 Transcription Factor/deficiency , Space Perception/drug effects , Animals , CREB-Binding Protein/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Laminin/metabolism , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Knockout , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , STAT1 Transcription Factor/genetics , Space Perception/physiology
11.
J Biol Chem ; 284(7): 4073-89, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19088076

ABSTRACT

Serum- and glucocorticoid-inducible kinase 1 (SGK1) is a downstream target of phosphatidylinositol 3-kinase signaling, and it regulates various cellular and physiological functions, but the SGK1 substrate proteins and genes regulated by SGK1 are less known. Here we have identified IkappaB kinase alpha (IKKalpha) as a novel substrate of SGK1 by using biochemical and bioinformatic approaches. SGK1 directly phosphorylates IKKalpha at Thr-23 and indirectly activates IKKalpha at Ser-180. Furthermore, SGK1 enhanced nuclear factor kappaB (NF-kappaB) activity and up-regulated N-methyl-d-aspartate receptor NR2A and NR2B expression through activation of IKKalpha at Thr-23 and Ser-180, and these two residues play an equally important role in mediating these effects of SGK1. Although SGK1 does not phosphorylate IKKbeta, IKKbeta activity is still required for IKK complex activation and for SGK1 phosphorylation and activation of NF-kappaB. In addition, SGK1 increased the acetylation of NF-kappaB through phosphorylation of p300 at Ser-1834, and this also leads to NF-kappaB activation and NR2A and NR2B expression. Moreover, an endogenous stimulus of SGK1, insulin, increased IKKalpha and NF-kappaB phosphorylation as well as NF-kappaB acetylation and NF-kappaB activity, but SGK1 small interfering RNA transfection blocked these effects of insulin. In examination of the functional significance of the SGK1-IKKalpha-NF-kappaB signaling pathway, we found that transfection of the IKKalpha double mutant (IKKalphaT23A/S180A) to rat hippocampus antagonized SGK-1-mediated spatial memory facilitation. Our results together demonstrated novel substrate proteins of SGK1 and novel SGK1 signaling pathways. Activation of these signaling pathways enhances NR2A and NR2B expression that is implicated in neuronal plasticity.


Subject(s)
E1A-Associated p300 Protein/metabolism , Hippocampus/metabolism , I-kappa B Kinase/metabolism , Immediate-Early Proteins/metabolism , NF-kappa B/metabolism , Neuronal Plasticity/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Up-Regulation/physiology , Animals , E1A-Associated p300 Protein/genetics , Humans , I-kappa B Kinase/genetics , Immediate-Early Proteins/genetics , NF-kappa B/genetics , PC12 Cells , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/physiology
12.
J Neurochem ; 105(3): 820-32, 2008 May.
Article in English | MEDLINE | ID: mdl-18088355

ABSTRACT

Serum- and glucocorticoid-inducible kinase 1 (SGK1) has been shown to play an important role in spatial memory formation, but the molecular mechanism underlying this effect of SGK1 was not known. zif268 is an immediate early gene that is induced by water maze learning. To investigate the role of SGK1 in the regulation of zif268 expression, the dominant negative mutant of SGK1, SGK1 S422A, was infused to the hippocampal CA1 area of rats, and was found to decrease significantly the mRNA level of zif268 in both naïve animals and trained animals. SGK1 was also found to phosphorylate serum response factor (SRF) at Ser73, Ser75, and Ser99, and phosphorylate CREB1 at Ser133. Inhibition of SGK1 phosphorylation sites on SRF and CREB1 with alanine substitution significantly diminished SGK1-enhanced zif268 expression in the promoter-luciferase assay. SGK1 also phosphorylates Elk-1 and SGK1 phosphorylation of Elk-1 decreased the transcriptional activity of Elk-1. But SGK1 phosphorylation of Elk-1 did not affect SGK1-enhanced zif268 expression. Moreover, the phosphorylation of SGK1 was increased in rat CA1 area after water maze learning, accompanied by increased phosphorylation of SRF at Ser99 and increased phosphorylation of CREB1 at Ser133. All these effects were antagonized by SGK1 S422A transfection. These results together suggest that SGK1 enhances zif268 expression through the mediation of SRF and CREB1, and these signaling pathways are associated with spatial memory formation in rats.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 1/genetics , Hippocampus/metabolism , Immediate-Early Proteins/genetics , Memory/physiology , Protein Serine-Threonine Kinases/genetics , Animals , Binding Sites/physiology , Cell Line , Down-Regulation/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Regulation/physiology , Humans , Immediate-Early Proteins/metabolism , Maze Learning/physiology , PC12 Cells , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , Rats , Serine/metabolism , Serum Response Factor/metabolism , Space Perception/physiology , ets-Domain Protein Elk-1/metabolism
13.
Neurobiol Aging ; 24(1): 105-16, 2003.
Article in English | MEDLINE | ID: mdl-12493556

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is a specific neurotrophic factor for midbrain dopamine (DA) neurons, but the mechanism underlying the neurotrophic action of GDNF is not well known. The cell adhesion molecules integrin and Neural cell adhesion molecule (NCAM) play important roles in neurite outgrowth and fasciculation. In the present study, we found that subchronic GDNF administration to the pars compacta of substantia nigra in rats increased the expression of integrin alphav and NCAM. Immunostaining results demonstrated the wide distribution of integrin alphav and NCAM in all mesencephalic neurons. The results also demonstrated the co-expression of TH with integrin alphav and NCAM in the same neurons of mesencephalic culture. Further, GDNF significantly increased integrin alphav expression in single TH-positive neurons. Function-blocking anti-integrin alphav and anti-NCAM antibodies antagonized the effects of GDNF on DA neuron survival, outgrowth, DA turnover, and locomotor activity in rats. These results demonstrate that integrin alphav and NCAM mediate the effects of GDNF on DA neuron survival and outgrowth during development and on DA turnover and motor function during adulthood.


Subject(s)
Dopamine/metabolism , Integrin alphaV/physiology , Motor Activity/drug effects , Nerve Growth Factors/pharmacology , Neural Cell Adhesion Molecules/physiology , Neurons/drug effects , Analysis of Variance , Animals , Antibodies/pharmacology , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Glial Cell Line-Derived Neurotrophic Factor , Immunohistochemistry , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/metabolism , Motor Activity/physiology , Nerve Growth Factors/administration & dosage , Neurons/physiology , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...