Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 159
Filter
Add more filters










Publication year range
1.
Genes Brain Behav ; 15(3): 348-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26749572

ABSTRACT

Abnormally active glycogen synthase kinase-3 (GSK3) contributes to pathological processes in multiple psychiatric and neurological disorders. Modeled in mice, this includes increasing susceptibility to dysregulation of mood-relevant behaviors, impairing performance in several cognitive tasks and impairing adult hippocampal neural precursor cell (NPC) proliferation. These deficits are all evident in GSK3α/ß knockin mice, in which serine-to-alanine mutations block the inhibitory serine phosphorylation regulation of both GSK3 isoforms, leaving GSK3 hyperactive. It was unknown if both GSK3 isoforms perform redundant actions in these processes, or if hyperactivity of one GSK3 isoform has a predominant effect. To test this, we examined GSK3α or GSK3ß knockin mice in which only one isoform was mutated to a hyperactive form. Only GSK3ß, not GSK3α, knockin mice displayed heightened vulnerability to the learned helplessness model of depression-like behavior. Three cognitive measures impaired in GSK3α/ß knockin mice showed differential regulation by GSK3 isoforms. Novel object recognition was impaired in GSK3ß, not in GSK3α, knockin mice, whereas temporal order memory was not impaired in GSK3α or GSK3ß knockin mice, and co-ordinate spatial processing was impaired in both GSK3α and GSK3ß knockin mice. Adult hippocampal NPC proliferation was severely impaired in GSK3ß knockin mice, but not impaired in GSK3α knockin mice. Increased activity of GSK3ß, in the absence of overexpression or disease pathology, is sufficient to impair mood regulation, novel object recognition and hippocampal NPC proliferation, whereas hyperactive GSK3α individually does not impair these processes. These results show that hyperactivity of the two GSK3 isoforms execute non-redundant effects on these processes.


Subject(s)
Depression/enzymology , Glycogen Synthase Kinase 3/metabolism , Hippocampus/enzymology , Memory/physiology , Animals , Cell Proliferation/physiology , Depression/pathology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , Male , Mice , Mice, Transgenic , Phosphorylation , Protein Isoforms
2.
Transl Psychiatry ; 4: e488, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25514751

ABSTRACT

Suicide is one of the leading causes of death in the United States, yet it remains difficult to understand the mechanistic provocations and to intervene therapeutically. Stress is recognized as a frequent precursor to suicide. Psychological stress is well established to cause activation of the inflammatory response, including causing neuroinflammation, an increase of inflammatory molecules in the central nervous system (CNS). Neuroinflammation is increasingly recognized as affecting many aspects of CNS functions and behaviors. In particular, much evidence demonstrates that inflammatory markers are elevated in traits that have been linked to suicidal behavior, including aggression, impulsivity and depression. Lithium is recognized as significantly reducing suicidal behavior, is anti-inflammatory and diminishes aggression, impulsivity and depression traits, each of which is associated with elevated inflammation. The anti-inflammatory effects of lithium result from its inhibition of glycogen synthase kinase-3 (GSK3). GSK3 has been demonstrated to strongly promote inflammation, aggressive behavior in rodents and depression-like behaviors in rodents, whereas regulation of impulsivity by GSK3 has not yet been investigated. Altogether, evidence is building supporting the hypothesis that stress activates GSK3, which in turn promotes inflammation, and that inflammation is linked to behaviors associated with suicide, including particularly aggression, impulsivity and depression. Further investigation of these links may provide a clearer understanding of the causes of suicidal behavior and provide leads for the development of effective preventative interventions, which may include inhibitors of GSK3.


Subject(s)
Aggression/physiology , Depression , Glycogen Synthase Kinase 3 , Impulsive Behavior/physiology , Inflammation , Lithium/pharmacology , Stress, Psychological , Suicide Prevention , Aggression/drug effects , Animals , Depression/drug therapy , Depression/etiology , Depression/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Impulsive Behavior/drug effects , Inflammation/drug therapy , Inflammation/etiology , Inflammation/metabolism , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/metabolism
3.
Genes Brain Behav ; 12(7): 723-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23941202

ABSTRACT

Fragile X syndrome (FXS) is caused by suppressed expression of fragile X mental retardation protein (FMRP), which results in intellectual disability accompanied by many variably manifested characteristics, such as hyperactivity, seizures and autistic-like behaviors. Treatment of mice that lack FMRP, Fmr1 knockout (KO) mice, with lithium has been reported to ameliorate locomotor hyperactivity, prevent hypersensitivity to audiogenic seizures, improve passive avoidance behavior and attenuate sociability deficits. To focus on the defining characteristic of FXS, which is cognitive impairment, we tested if lithium treatment ameliorated impairments in four cognitive tasks in Fmr1 KO mice, tested if the response to lithium differed in adolescent and adult mice and tested if therapeutic effects persisted after discontinuation of lithium administration. Fmr1 KO mice displayed impaired cognition in the novel object detection task, temporal ordering for objects task and coordinate and categorical spatial processing tasks. Chronic lithium treatment of adolescent (from 4 to 8 weeks of age) and adult (from 8 to 12 weeks of age) mice abolished cognitive impairments in all four cognitive tasks. Cognitive deficits returned after lithium treatment was discontinued for 4 weeks. These results show that Fmr1 KO mice exhibit severe impairments in these cognitive tasks, that lithium is equally effective in normalizing cognition in these tasks whether it is administered to young or adult mice and that lithium administration must be continued for the cognitive improvements to be sustained. These findings provide further evidence that lithium administration may be beneficial for individuals with FXS.


Subject(s)
Cognition/drug effects , Fragile X Syndrome/drug therapy , Lithium/pharmacology , Age Factors , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Lithium/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Neuroscience ; 226: 1-9, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22986169

ABSTRACT

Glycogen synthase kinase 3 (GSK3) is a serine/threonine kinase that has been implicated in psychiatric diseases, neurodevelopment, and circadian regulation. Both GSK3 isoforms, α and ß, exhibit a 24-h variation of inhibitory phosphorylation within the suprachiasmatic nucleus (SCN), the primary circadian pacemaker. We examined the hypothesis that rhythmic GSK3 activity is critical for robust circadian rhythmicity using GSK3α(21A/21A)/ß(9A/9A) knock-in mice with serine-alanine substitutions at the inhibitory phosphorylation sites, making both forms constitutively active. We monitored wheel-running locomotor activity of GSK3 knock-in mice and used loose-patch electrophysiology to examine the effect of chronic GSK3 activity on circadian behavior and SCN neuronal activity. Double transgenic GSK3α/ß knock-in mice exhibit disrupted behavioral rhythmicity, including significantly decreased rhythmic amplitude, lengthened active period, and increased activity bouts per day. This behavioral disruption was dependent on chronic activation of both GSK3 isoforms and was not seen in single transgenic GSK3α or GSK3ß knock-in mice. Underlying the behavioral changes, SCN neurons from double transgenic GSK3α/ß knock-in mice exhibited significantly higher spike rates during the subjective night compared to those from wild-type controls, with no differences detected during the subjective day. These results suggest that constitutive activation of GSK3 results in the loss of the typical day/night variation of SCN neuronal activity. Together, these results implicate GSK3 activity as a critical regulator of circadian behavior and neurophysiological rhythms. Because GSK3 has been implicated in numerous pathologies, understanding how GSK3 modulates circadian rhythms and neurophysiological activity may lead to novel therapeutics for pathological disorders and circadian rhythm dysfunction.


Subject(s)
Action Potentials/physiology , Circadian Rhythm/physiology , Glycogen Synthase Kinase 3/physiology , Suprachiasmatic Nucleus/physiology , Animals , Animals, Genetically Modified , Blotting, Western , Electrophysiological Phenomena , Enzyme Activation/physiology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Neurons/physiology , Patch-Clamp Techniques , Phosphorylation
6.
Neuroscience ; 169(3): 1063-70, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20553816

ABSTRACT

Inflammatory tolerance is the down-regulation of inflammation upon repeated stimuli, which is well-established to occur in peripheral immune cells. However, less is known about inflammatory tolerance in the brain although it may provide an important protective mechanism from detrimental consequences of prolonged inflammation, which appears to occur in many psychiatric and neurodegenerative conditions. Array analysis of 308 inflammatory molecules produced by mouse primary astrocytes after two sequential stimulations with lipopolysaccharide (LPS) distinguished three classes, tolerant, sensitized and unaltered groups. For many of these inflammatory molecules, inhibition of glycogen synthase kinase-3 (GSK3) increased tolerance and reduced sensitization. Focusing on LPS-tolerance in interleukin-6 (IL-6) production, we found that microglia exhibited a strong tolerance response that matched that of macrophages, whereas astrocytes exhibited only partial tolerance. The astrocyte semi-tolerance was found to be regulated by GSK3. GSK3 inhibitors or knocking down GSK3 levels promoted LPS-tolerance and astrocytes expressing constitutively active GSK3 did not develop LPS-tolerance. These findings identify the critical role of GSK3 in counteracting IL-6 inflammatory tolerance in cells of the CNS, supporting the therapeutic potential of GSK3 inhibitors to reduce neuroinflammation by promoting tolerance.


Subject(s)
Astrocytes/metabolism , Glycogen Synthase Kinase 3/physiology , Inflammation/metabolism , Animals , Astrocytes/drug effects , Cells, Cultured , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics
7.
Cell Death Differ ; 15(12): 1887-900, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18846110

ABSTRACT

Stimulation of death receptors activates the extrinsic apoptotic signaling pathway that leads to cell death. Although many steps of this apoptotic signaling cascade are known, few mechanisms that counterbalance the death signal have been described. We identified an antiapoptotic protein complex associated with death receptors that contains glycogen synthase kinase-3 (GSK3), DDX3 and cellular inhibitor of apoptosis protein-1 (cIAP-1). GSK3, DDX3 and cIAP-1 are associated in cells with each other and with death receptors. Blocking the actions of GSK3 or DDX3 potentiated caspase-3 activation induced by stimulation of four different death receptors in several types of cells. GSK3 restrained apoptotic signaling by inhibiting formation of the death-inducing signaling complex and caspase-8 activation. Stimulated death receptors surmount the antiapoptotic complex by causing GSK3 inactivation and cleavage of DDX3 and cIAP-1 to enable progression of the apoptotic signaling cascade, but the antiapoptotic complex remains functional in cancer cells resistant to death receptor stimulation, a resistance that is overcome by GSK3 inhibitors. Thus, an antiapoptotic complex of GSK3, DDX3 and cIAP-1 caps death receptors, providing a checkpoint to counterbalance apoptotic signaling.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Receptors, Death Domain/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Humans , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
8.
Mol Psychiatry ; 7 Suppl 1: S35-45, 2002.
Article in English | MEDLINE | ID: mdl-11986994

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) is a central figure in many intracellular signaling systems and is directly regulated by lithium. Substantial evidence now indicates that an important property of the mood stabilizer, lithium, is to influence GSK3beta-linked signaling pathways. This raises the possibility that other mood stabilizers act in a similar manner, which may include modulation of signaling systems leading to GSK3beta, direct regulation of GSK3beta or regulation of signaling intermediates downstream of GSK3beta. Downstream targets of GSK3beta, and thus potential targets of mood stabilizers, are several key transcription factors, including beta-catenin, AP-1, cyclic AMP-response element binding protein, NFkappaB, Myc, heat shock factor-1, nuclear factor of activated T-cells and CCAAT/enhancer-binding proteins. GSK3beta also is an important modulator of cell death, which may be a consequence of its regulatory effects on transcription factor activities. GSK3beta facilitates apoptosis, and lithium's inhibition of GSK3beta supports cell survival. Thus, signaling systems determining cell fate appear to be important targets of mood stabilizers, and these may include signaling pathways encompassing GSK3beta, including transcription factors regulated by GSK3beta.


Subject(s)
Antidepressive Agents/therapeutic use , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Microtubule-Associated Proteins/metabolism , Mood Disorders/drug therapy , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Survival/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Models, Neurological , Neuroprotective Agents/pharmacology
9.
Brain Res ; 919(1): 106-14, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11689167

ABSTRACT

The compound 1-methyl-4-phenylpyridinium (MPP) is a selective inhibitor of mitochondrial complex I, and is widely used in model systems to elicit neurochemical alterations that may be associated with Parkinson's disease. In the present study treatment of human neuroblastoma SH-SY5Y cells with MPP resulted in a time- and concentration-dependent activation of the apoptosis-associated cysteine protease caspase-3, and caused morphological changes characteristic of apoptosis. To test if the activation state of the cell survival-promoting phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway affects MPP-induced caspase-3 activation, PI3K was inhibited with LY294002, or activated with insulin-like growth factor-1. MPP-induced caspase-3 activation was increased by inhibition of PI3K, and decreased by stimulation of PI3K, indicative of anti-apoptotic signaling by the PI3K/Akt pathway. To test if glycogen synthase kinase-3beta (GSK3beta), a pro-apoptotic kinase that is inhibited by Akt, is involved in regulating MPP-induced apoptosis, overexpression of GSK3beta and lithium, a selective inhibitor of GSK3beta, were used to directly alter GSK3beta activity. MPP-induced caspase-3 activity was increased by overexpression of GSK3beta. Conversely, the GSK3beta inhibitor lithium attenuated MPP-induced caspase-3 activation. To test if these regulatory interactions applied to other mitochondrial complex I inhibitors, cells were treated with rotenone. Rotenone-induced activation of caspase-3 was enhanced by inhibition of PI3K or increased GSK3beta activity, and was attenuated by inhibiting GSK3beta with lithium. Overall, these results indicate that inhibition of GSK3beta provides protection against the toxic effects of agents, such as MPP and rotenone, that impair mitochondrial function.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Caspases/metabolism , Lithium/pharmacology , Mitochondria/drug effects , Mitochondria/enzymology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/pharmacology , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Caspase 3 , Chromones/pharmacology , Drug Synergism , Electron Transport Complex I , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Herbicides/pharmacology , Humans , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Rotenone/pharmacology , Tumor Cells, Cultured
10.
J Neurochem ; 78(6): 1219-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11579131

ABSTRACT

The regulatory influences of glycogen synthase kinase-3 beta (GSK3 beta) and lithium on the activity of cyclic AMP response element binding protein (CREB) were examined in human neuroblastoma SH-SY5Y cells. Activation of Akt (protein kinase B) with serum-increased phospho-serine-9-GSK3 beta (the inactive form of the enzyme), inhibited GSK3 beta activity, and increased CREB DNA binding activity. Inhibition of GSK3 beta by another paradigm, treatment with the selective inhibitor lithium, also increased CREB DNA binding activity. The inhibitory regulation of CREB DNA binding activity by GSK3 beta also was evident in differentiated SH-SY5Y cells, indicating that this regulatory interaction is maintained in non-proliferating cells. These results demonstrate that inhibition of GSK3 beta by serine-9 phosphorylation or directly by lithium increases CREB activation. Conversely, overexpression of active GSK3 beta to 3.5-fold the normal levels completely blocked increases in CREB DNA binding activity induced by epidermal growth factor, insulin-like growth factor-1, forskolin, and cyclic AMP. The inhibitory effects due to overexpressed GSK3 beta were reversed by treatment with lithium and with another GSK 3beta inhibitor, sodium valproate. Overall, these results demonstrate that GSK3 beta inhibits, and lithium enhances, CREB activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , DNA/metabolism , Lithium/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA/antagonists & inhibitors , Drug Synergism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Tumor Cells, Cultured
11.
Neurochem Res ; 26(7): 809-16, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11565612

ABSTRACT

Muscarinic receptor-mediated changes in protein tyrosine phosphorylation were examined in differentiated human neuroblastoma SH-SY5Y cells. Treatment of differentiated cells with 1 mM carbachol caused rapid increases in the tyrosine phosphorylation of focal adhesion kinase (FAK), Cas, and paxillin. The src family kinase-selective inhibitor PP1 reduced carbachol-stimulated tyrosine phosphorylation of FAK, Cas, and paxillin by 50 to 75%. In contrast, carbachol-stimulated activation of ERK1/2 was unaffected by PP1. Src family kinase activation by carbachol was further demonstrated by increased carbachol-induced tyrosine phosphorylation of the src-substrate, p120, and tyrosine phosphorylation of the src family kinase activation-associated autophosphorylation site. Site-specific FAK phosphotyrosine antibodies were used to determine that the carbachol-stimulated increase in the autophosphorylation of FAK was unaffected by pretreatment with PP1, whereas the carbachol-stimulated increase in the src family kinase-mediated phosphotyrosine of FAK was completely blocked by pretreatment with PP1. In SH-SY5Y cell lines stably overexpressing Fyn, the phosphotyrosine immunoreactivity of FAK was 625% that of control cells. Thus, muscarinic receptors activate protein tyrosine phosphorylation in differentiated cells, and the tyrosine phosphorylation of FAK, Cas, and paxillin, but not ERK1/2, is mediated by a src family tyrosine kinase activated in response to stimulation of muscarinic receptors.


Subject(s)
Receptors, Muscarinic/physiology , Tyrosine/metabolism , src-Family Kinases/physiology , Carbachol/pharmacology , Cell Differentiation , Cellular Apoptosis Susceptibility Protein/metabolism , Cholinergic Agonists/pharmacology , Cytoskeletal Proteins/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Neurons/pathology , Neurons/physiology , Paxillin , Phenotype , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Tumor Cells, Cultured/pathology
12.
Prog Neurobiol ; 65(4): 391-426, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11527574

ABSTRACT

Glycogen synthase kinase-3beta (GSK3beta) is a fascinating enzyme with an astoundingly diverse number of actions in intracellular signaling systems. GSK3beta activity is regulated by serine (inhibitory) and tyrosine (stimulatory) phosphorylation, by protein complex formation, and by its intracellular localization. GSK3beta phosphorylates and thereby regulates the functions of many metabolic, signaling, and structural proteins. Notable among the signaling proteins regulated by GSK3beta are the many transcription factors, including activator protein-1, cyclic AMP response element binding protein, heat shock factor-1, nuclear factor of activated T cells, Myc, beta-catenin, CCAAT/enhancer binding protein, and NFkappaB. Lithium, the primary therapeutic agent for bipolar mood disorder, is a selective inhibitor of GSK3beta. This raises the possibility that dysregulation of GSK3beta and its inhibition by lithium may contribute to the disorder and its treatment, respectively. GSK3beta has been linked to all of the primary abnormalities associated with Alzheimer's disease. These include interactions between GSK3beta and components of the plaque-producing amyloid system, the participation of GSK3beta in phosphorylating the microtubule-binding protein tau that may contribute to the formation of neurofibrillary tangles, and interactions of GSK3beta with presenilin and other Alzheimer's disease-associated proteins. GSK3beta also regulates cell survival, as it facilitates a variety of apoptotic mechanisms, and lithium provides protection from many insults. Thus, GSK3beta has a central role regulating neuronal plasticity, gene expression, and cell survival, and may be a key component of certain psychiatric and neurodegenerative diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Physiological Phenomena , Signal Transduction/physiology , Alzheimer Disease/physiopathology , Animals , Bipolar Disorder/physiopathology , Gene Expression Regulation, Enzymologic/physiology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Transcription Factors/metabolism
13.
Arch Biochem Biophys ; 392(2): 192-6, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11488592

ABSTRACT

RGS2, a regulators of G-protein signaling family member, regulates G-protein signaling and is itself controlled in part by regulated expression. We tested if cell stress regulates RGS2 expression in human astrocytoma 1321N1 cells. Treatment with H2O2 increased RGS2 mRNA levels time- and concentration-dependently, with 200 microM H2O2 causing an approximately eightfold increase after 2 h. Peroxynitrite and heat shock also increased RGS2 mRNA levels. H2O2-induced RGS2 expression was negatively regulated by phosphoinositide-3-kinase and extracellular signal-regulated kinases. H2O2 also concentration-dependently increased RGS2 protein levels, and the RGS2 appeared to be predominantly in the nucleus. These results demonstrate that RGS2 expression is up-regulated by cell stress.


Subject(s)
Hot Temperature , Oxidative Stress , Astrocytoma/metabolism , Blotting, Northern , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Humans , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RGS Proteins/chemistry , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Tumor Cells, Cultured , Up-Regulation
14.
J Biol Chem ; 276(40): 37436-42, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11495916

ABSTRACT

The goal of this study was to determine whether the intracellular distribution of the proapoptotic enzyme glycogen synthase kinase-3 beta (GSK-3 beta) is dynamically regulated by conditions that activate apoptotic signaling cascades. In untreated human neuroblastoma SH-SY5Y cells, GSK-3 beta was predominantly cytosolic, although a low level was also detected in the nucleus. The nuclear level of GSK-3 beta was rapidly increased after exposure of cells to serum-free media, heat shock, or staurosporine. Although each of these conditions caused changes in the serine 9 and/or tyrosine phosphorylation of GSK-3 beta, neither of these modifications was correlated with nuclear accumulation of GSK-3 beta. Heat shock and staurosporine treatments increased nuclear GSK-3 beta prior to activation of caspase-9 and caspase-3, and this nuclear accumulation of GSK-3 beta was unaltered by pretreatment with a general caspase inhibitor. The GSK-3 beta inhibitor lithium did not alter heat shock-induced nuclear accumulation of GSK-3 beta but increased the nuclear level of cyclin D1, indicating that cyclin D1 is a substrate of nuclear GSK-3 beta. Thus, the intracellular distribution of GSK-3 beta is dynamically regulated by signaling cascades, and apoptotic stimuli cause increased nuclear levels of GSK-3 beta, which facilitates interactions with nuclear substrates.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Nucleus/drug effects , Flavoproteins/pharmacology , Membrane Proteins/pharmacology , Apoptosis Inducing Factor , Cell Nucleus/enzymology , Cyclin D1/metabolism , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Hot Temperature , Humans , Lithium/pharmacology , Phosphorylation , Serine/metabolism , Staurosporine/pharmacology , Tumor Cells, Cultured , Tyrosine/metabolism
15.
Brain Res ; 903(1-2): 226-30, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11382407

ABSTRACT

This study examined the effects of overexpression of presenilin-1 wild-type (PS1wt) or mutant L286V (PS1m) in human neuroblastoma SH-SY5Y cells on signal transduction systems. Oxotremorine-M-induced activation of AP-1 was 40--53% lower in PS1wt than control cells, and further impaired (63--76%) in PS1m cells. Heat shock (45 degrees C) activated Akt, increased heat shock factor-1 (HSF-1) DNA binding activity, and increased levels of heat shock protein 70, and these responses were not altered by overexpression of PS1wt or PS1m. H(2)O(2) also caused a time-dependent increase in HSF-1 DNA binding activity which was similar in all cell lines. Thus, overexpression of PS1wt reduced muscarinic receptor-mediated activation of AP-1, and PS1m overexpression caused greater inhibition, but stress-induced activation of Akt and HSF-1 was unaffected by either PS1wt or PS1m.


Subject(s)
Acetylcholine/physiology , Heat-Shock Response/physiology , Membrane Proteins/genetics , Oxidative Stress/physiology , Oxotremorine/analogs & derivatives , Signal Transduction/physiology , Gene Expression/physiology , Humans , Membrane Proteins/metabolism , Muscarinic Agonists/pharmacology , Neuroblastoma , Oxotremorine/pharmacology , Presenilin-1 , Receptors, Muscarinic/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
16.
Biochem Biophys Res Commun ; 283(1): 102-6, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11322774

ABSTRACT

RGS2, a Regulators of G-protein Signaling family member, regulates signaling activities of G-proteins, and RGS2 itself is controlled in part by regulation of its expression. This investigation extended previous studies of the regulation of RGS2 expression by examining the effects of stress, differentiation, and signaling activities on RGS2 mRNA level in human neuroblastoma SH-SY5Y cells. Cell stress induced by heat shock rapidly and transiently increased RGS2 mRNA levels, whereas differentiation to a neuronal phenotype reduced basal RGS2 mRNA levels by 50%. RGS2 mRNA levels were increased in differentiated cells by heat shock, carbachol, and activation of protein kinase C. After transient transfection of GFP-tagged RGS2, a predominant nuclear localization was observed by confocal microscopy. Thus, RGS2 expression is regulated by stress and differentiation, as well as by second messenger signaling, and transfected GFP-RGS2 is predominantly nuclear.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Neurons/metabolism , RGS Proteins/metabolism , Carbachol/pharmacology , Cell Differentiation/drug effects , Cell Line , Cholinergic Agonists/pharmacology , Colforsin/pharmacology , Culture Media/pharmacology , Cyclic AMP/metabolism , Green Fluorescent Proteins , Heat-Shock Response/physiology , Humans , Luminescent Proteins/genetics , Neuroblastoma/metabolism , Neurons/cytology , Neurons/drug effects , Protein Kinase C/metabolism , RGS Proteins/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transfection
17.
Biochim Biophys Acta ; 1541(3): 201-11, 2001 Dec 19.
Article in English | MEDLINE | ID: mdl-11755214

ABSTRACT

Regulators of G-protein Signaling (RGS) proteins attenuate signaling activities of G proteins, and modulation of expression appears to be a primary mechanism for regulating RGS proteins. In human astrocytoma 1321N1 cells RGS2 expression was increased by activation of muscarinic receptors coupled to phosphoinositide signaling with carbachol, or by increased cyclic AMP production, demonstrating that both signaling systems can increase the expression of a RGS family member in a single cell type. Carbachol-stimulated increases in endogenous RGS2 protein levels appeared by immunocytochemical analysis to be largely confined to the nucleus, and this localization was confirmed by Western blot analysis which showed increased nuclear, but not cytosolic, RGS2 after carbachol treatment. Additionally, transiently expressed green fluorescent protein (GFP)-tagged, 6xHis-tagged, or unmodified RGS2 resulted in a predominant nuclear localization, as well as a distinct accumulation of RGS2 along the plasma membrane. The intranuclear localization of GFP-RGS2 was confirmed with confocal microscopy. Thus, RGS2 expression is rapidly and transiently increased by phosphoinositide signaling and by cyclic AMP, and endogenous and transfected RGS2 is largely, although not entirely, localized in the nucleus.


Subject(s)
Cell Nucleus/metabolism , RGS Proteins/biosynthesis , Second Messenger Systems/physiology , Astrocytoma , Blotting, Western , Carbachol/pharmacology , Cell Membrane/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cytosol/metabolism , Humans , Immunohistochemistry , Isoproterenol/pharmacology , Microscopy, Confocal , RGS Proteins/analysis , RGS Proteins/genetics , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Adrenergic, beta/drug effects , Receptors, Muscarinic/drug effects , Time Factors , Transfection , Tumor Cells, Cultured
18.
J Neurochem ; 75(6): 2401-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11080191

ABSTRACT

Elevated temperatures activate the survival promoters Akt and heat shock factor-1 (HSF-1), a transcription factor that induces the expression of heat shock proteins (HSPs), such as HSP-70. Because neuronal mechanisms controlling these responses are not known, these were investigated in human neuroblastoma SH-SY5Y cells. Heat shock (45 degrees C) rapidly activated Akt, extracellular signal-regulated kinases 1 and 2 (ERK1/2), and p38, but only Akt was activated in a phosphatidylinositol 3-kinase (PI-3K)-dependent manner, as the PI-3K inhibitors LY294002 and wortmannin blocked Akt activation, but not ERK1/2 or p38 activation. Akt activation was not blocked by inhibition of p38 or ERK1/2, indicating the independence of these signaling systems. Heat shock treatment also caused a rapid increase in HSF-1 DNA binding activity that was partially dependent on PI-3K activity, as both the PI-3K inhibitors attenuated this response. Because Akt inhibits glycogen synthase kinase-3beta (GSK-3beta), an enzyme that facilitates cell death, we tested if GSK-3beta is a negative regulator of HSF-1 activation. Overexpression of GSK-3beta impaired heat shock-induced activation of HSF-1, and also reduced HSP-70 production, which was partially restored by the GSK-3beta inhibitor lithium. Thus, heat shock-induced activation of PI-3K and the inhibitory effect of GSK-3beta on HSF-1 activation and HSP-70 expression imply that Akt-induced inhibition of GSK-3beta contributes to the activation of HSF-1.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/genetics , DNA/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , HSP70 Heat-Shock Proteins/biosynthesis , Heat Shock Transcription Factors , Heat-Shock Response/drug effects , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Transcription Factors , Transfection , p38 Mitogen-Activated Protein Kinases
19.
Neurobiol Aging ; 21(1): 31-8, 2000.
Article in English | MEDLINE | ID: mdl-10794846

ABSTRACT

Alzheimer's disease cybrid cells produced by replacing endogenous mitochondria in human neuroblastoma SH-SY5Y cells with platelet mitochondria from subjects with Alzheimer's disease have higher levels of reactive oxygen species than do cybrid cells with mitochondria from control subjects. These cells were used to test if this chronic mild increase in reactive oxygen species affects muscarinic receptor-coupled signaling activities. Basal and carbachol-stimulated phosphoinositide hydrolysis were higher, and there was less inhibition by glutathione depletion, in Alzheimer's disease than control cybrid cells. Elevated phosphoinositide hydrolysis in Alzheimer's disease cybrid cells also was evident upon direct activation of G-proteins (Gq/11) linked to phosphoinositide signaling or of phospholipase C, but immunoblot analyses revealed equivalent levels of Gq/11 and phospholipase C in both cell lines. These results indicate that there is up-regulation of phosphoinositide signaling in Alzheimer's disease cybrid cells in association with chronic mild oxidative stress, although treatment of cells with H(2)O(2) to induce greater acute oxidative stress caused decreases in carbachol-stimulated phosphoinositide hydrolysis that were similar in Alzheimer's disease and control cybrid cells. In contrast to phosphoinositide hydrolysis, carbachol-stimulated AP-1 DNA binding activity was lower in Alzheimer's disease than control cybrid cells, and this deficit was associated with deficient protein kinase C-mediated activation of AP-1. Overall, these results demonstrate that chronically elevated reactive oxygen species in Alzheimer's disease cybrid cells are associated with a more robust phosphoinositide signaling system, but lower signaling to activation of AP-1. These alterations may represent adaptations to exposure to oxidants, which precede more widespread deficits in signaling associated with more severe oxidative stress.


Subject(s)
Alzheimer Disease/metabolism , Hybrid Cells/metabolism , Phosphatidylinositols/metabolism , Receptors, Muscarinic/metabolism , Transcription Factor AP-1/metabolism , Alzheimer Disease/pathology , Buthionine Sulfoximine/pharmacology , Carbachol/pharmacology , Cells, Cultured , Cholinergic Agonists/pharmacology , DNA/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , Glutathione/metabolism , Humans , Hybrid Cells/cytology , Hydrogen Peroxide/pharmacology , Hydrolysis/drug effects , Mitochondria/metabolism , Mitochondria/transplantation , Oxidative Stress/drug effects , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Type C Phospholipases/metabolism
20.
Arch Biochem Biophys ; 376(2): 365-70, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10775424

ABSTRACT

Although peroxynitrite appears to contribute to neuronal dysfunction in several neurodegenerative disorders, little is known about how peroxynitrite affects cellular signaling processes. This study investigated if peroxynitrite affects the mitogen-activated protein kinases, extracellular-regulated kinases 1 and 2 (ERK1/2) and p38. Exposure of PC12 cells to 500 microM peroxynitrite activated ERK1/2 and p38 within 5 min and this was followed by gradual decreases in activation over the next 25 min. Activation of ERK1/2 by peroxynitrite was mediated by activation of the epidermal growth factor (EGF) receptor in a calcium/calmodulin-dependent kinase II- and src family tyrosine kinase-dependent manner, as it was blocked by the selective EGF receptor inhibitor AG1478, by KN62, an inhibitor of calcium/calmodulin-dependent kinase II, and by PP1, a src family tyrosine kinase inhibitor. Activation of p38 by peroxynitrite was independent of the EGF receptor, required activation of calcium/calmodulin-dependent kinase II and src family tyrosine kinases, and was modulated by nerve growth factor (NGF) in a time-dependent manner. Pretreatment with NGF (2 h) attenuated, whereas cotreatment with NGF potentiated, peroxynitrite-induced activation of p38. Thus, peroxynitrite activates ERK1/2 and p38, activation of EGF receptors, calcium/calmodulin-dependent kinase II, and src family tyrosine kinases participate in these signaling responses to peroxynitrite, and peroxynitrite- and NGF-induced signaling activities converge on p38.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitrates/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Nerve Growth Factor/pharmacology , Nitrates/antagonists & inhibitors , PC12 Cells , Phosphorylation/drug effects , Quinazolines , Rats , Tyrphostins/pharmacology , p38 Mitogen-Activated Protein Kinases , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...