Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 516(3): 976-982, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31277940

ABSTRACT

Actin is a highly abundant cytoskeletal protein that is essential for all eukaryotic cells and participates in many structural and functional roles. It has long been noted that estrogen affects cellular morphology. However, recent studies observed that both estrogen and tamoxifen induce a remarkable cytoskeletal remodeling independent of ER. In addition to ER, G protein-coupled estrogen receptor 1 (GPER, also known as GPR30) also binds to estrogen with high affinity and mediates intracellular estrogenic signaling. Here, we show that activation of GPER by its specific agonist G-1 induces re-organization of F-actin cytoskeleton. We further demonstrate that GPER acts through PLCß-PKC and Rho/ROCK-LIMK-Cofilin pathway, which are upstream regulators of F-actin cytoskeleton assembly, thereby enhancing TAZ nuclear localization and activation. Furthermore, we find that LIMK1/2 is critical for GPER activation-induced breast cancer cell migration. Together, our results suggest that GPER mediates G-1-induced cytoskeleton assembly and GPER promotes breast cancer cell migration via PLCß-PKC and Rho/ROCK-LIMK-Cofilin pathway.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/genetics , Gene Expression Regulation, Neoplastic , Lim Kinases/genetics , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/ultrastructure , Actin Depolymerizing Factors/genetics , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclopentanes/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Lim Kinases/antagonists & inhibitors , Lim Kinases/metabolism , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Phospholipase C beta/genetics , Phospholipase C beta/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
2.
Mol Neurobiol ; 53(2): 983-994, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25575678

ABSTRACT

Endoplasmic reticulum (ER) stress has been indicated in the early stage of Alzheimer's disease (AD), in which tau hyperphosphorylation is one major pathological alteration. The elevation of binding immunoglobulin protein (Bip), an important ER chaperon, was reported in AD brain. It is important to study the roles of ER-related chaperons in tau hyperphosphorylation. In this research, increased Bip was found in the brains of the AD model mice (Tg2576) compared to the age-matched control mice. Meanwhile, deficiency of SIL1, an important co-chaperon of Bip, was observed in brains of Tg2576 mice and in ER stress both in vivo and in vitro. Then, we transfected Bip-EGFP plasmid into HEK293 cells stably expressing the longest human tau (HEK293/tau) or N2a cells and found that increased Bip induced tau hyperphosphorylation via activating glycogen synthase kinase-3ß (GSK-3ß), an important tau kinase, and increased the association with tau and GSK-3ß. When we overexpressed SIL1 in Bip-transfected HEK293/tau cells and thapsigargin-treated HEK293/tau cells, significantly reduced tau hyperphosphorylation and GSK-3ß activation were observed. These results suggested the important roles of ER-related chaperons, Bip and SIL1, in AD-like tau hyperphosphorylation.


Subject(s)
Endoplasmic Reticulum Stress , Guanine Nucleotide Exchange Factors/metabolism , Heat-Shock Proteins/metabolism , tau Proteins/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Indoles/pharmacology , Maleimides/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phosphorylation/drug effects , Rats, Sprague-Dawley , Thapsigargin/pharmacology
3.
Proc Natl Acad Sci U S A ; 111(46): 16586-91, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25378699

ABSTRACT

Intracellular accumulation of the abnormally modified tau is hallmark pathology of Alzheimer's disease (AD), but the mechanism leading to tau aggregation is not fully characterized. Here, we studied the effects of tau SUMOylation on its phosphorylation, ubiquitination, and degradation. We show that tau SUMOylation induces tau hyperphosphorylation at multiple AD-associated sites, whereas site-specific mutagenesis of tau at K340R (the SUMOylation site) or simultaneous inhibition of tau SUMOylation by ginkgolic acid abolishes the effect of small ubiquitin-like modifier protein 1 (SUMO-1). Conversely, tau hyperphosphorylation promotes its SUMOylation; the latter in turn inhibits tau degradation with reduction of solubility and ubiquitination of tau proteins. Furthermore, the enhanced SUMO-immunoreactivity, costained with the hyperphosphorylated tau, is detected in cerebral cortex of the AD brains, and ß-amyloid exposure of rat primary hippocampal neurons induces a dose-dependent SUMOylation of the hyperphosphorylated tau. Our findings suggest that tau SUMOylation reciprocally stimulates its phosphorylation and inhibits the ubiquitination-mediated tau degradation, which provides a new insight into the AD-like tau accumulation.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Point Mutation , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , tau Proteins/metabolism , Alzheimer Disease/pathology , Amino Acid Substitution , Amyloid beta-Peptides/pharmacology , Androstadienes/pharmacology , Animals , Cerebral Cortex/pathology , Female , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Indoles/pharmacology , Male , Maleimides/pharmacology , Middle Aged , Mutagenesis, Site-Directed , Mutation, Missense , Nerve Tissue Proteins/genetics , Peptide Fragments/pharmacology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Recombinant Fusion Proteins/metabolism , SUMO-1 Protein/genetics , Salicylates/pharmacology , Solubility , Sumoylation , Ubiquitination , Wortmannin , tau Proteins/genetics
4.
Int J Neuropsychopharmacol ; 16(5): 1071-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22999489

ABSTRACT

Ovariectomy is known as 'surgical menopause' with decreased levels of oestrogen in female rodents and its reported risks and adverse effects include cognitive impairment. In the brain, oestrogen exerts effects through its receptors, oestrogen receptor α (ERα) and ß (ERß). However, the role of ERα or ERß in ovariectomy-induced cognitive impairment needs further investigation. Here, we observed that bilaterally ovariectomized 3-month-old rats showed obvious spatial learning and memory deficits in the Morris water maze with significant loss of neurons and synapses in the hippocampus. In addition to the rapid decline in serum oestradiol levels, the expression of ERα, but not ERß, was decreased in the hippocampus starting 1 wk after ovariectomy. Prompt 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT) treatment (1 mg/kg.d), an agonist of ERα, improved the spatial learning and memory ability of ovariectomized rats and rescued ovariectomy-induced neuron loss by up-regulating the level of BCLxl, an important anti-apoptosis protein. Furthermore, PPT treatment also improved ovariectomy-induced hippocampal synapse loss and up-regulated the levels of synaptic proteins (synapsin I, NR2A and GluR1) and the activates of CaMK Πα, ERK and Akt. Thus, these results demonstrated that ERα plays an important role in neuroprotection and that prompt ERα rescue is effective to improve hippocampal-dependent cognition deficit after long-term ovariectomy.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/etiology , Estrogen Receptor alpha/agonists , Ginsenosides/therapeutic use , Ovariectomy/adverse effects , Sapogenins/therapeutic use , Spatial Behavior/drug effects , Animals , Apoptosis/drug effects , Cognition Disorders/metabolism , Estradiol/blood , Female , Gene Expression Regulation/drug effects , Ginsenosides/pharmacology , Maze Learning/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Sapogenins/pharmacology , Silver Staining , Time Factors
5.
J Alzheimers Dis ; 29(4): 727-40, 2012.
Article in English | MEDLINE | ID: mdl-22460328

ABSTRACT

Hyperphosphorylated tau is the major component of intracellular neurofibrillary tangles, which is positively correlated with the cognitive decline in Alzheimer's disease (AD). The upstream factors leading to tau hyperphosphorylation are still not fully understood. Endoplasmic reticulum (ER) stress has been indicated in AD pathogenesis and the increased level of binding immunoglobulin protein (Bip), an important ER associated chaperon, is increased in AD brain. Here hyperphosphorylation of tau, activation of glycogen synthase kinase-3ß (GSK-3ß), and elevation of Bip were induced by ventricular infusion of ER stressors, tunicamycin (TM) and thapsigargin (TG), in rats. GSK-3ß was found to be responsible for tau hyperphosphorylation induced by ER stressors both in vivo and in vitro. In addition, inhibited Akt, protein tyrosine phosphatase 1B, and activated Fyn were detected in vivo. Down-regulating Bip by tranfecting its siRNA plasmid significantly revised tau hyperphosphorylation in TG treated HEK293/tau cells, but the activation of GSK-3ß was still observed. By immunoprecipitation, we found that the binding levels of Bip to tau and GSK-3ß were significantly increased with the elevation of Bip in TM-treated rats. Moreover, in Bip overexpressed HEK293/tau cells, the binding levels of Bip to tau (mainly phosphorylated tau) and GSK-3ß were also significantly increased. However, ß-catenin, another important substrate of GSK-3ß, was not found bound to the increased Bip. All these data suggest an essential role of Bip in GSK-3ß dependent tau hyperphosphorylation in ER stress by promoting the binding of GSK-3ß to tau.


Subject(s)
Down-Regulation/physiology , Endoplasmic Reticulum Stress/physiology , Glycogen Synthase Kinase 3/metabolism , Lymphokines/metabolism , tau Proteins/metabolism , Animals , Cell Line, Transformed , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/genetics , Humans , Immunoprecipitation , Injections, Intraventricular , Lymphokines/genetics , Male , Mutation/genetics , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Thapsigargin/pharmacology , Time Factors , Transfection , Tunicamycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL