Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
Add more filters

Publication year range
1.
Med Sci Monit ; 25: 4627-4638, 2019 Jun 22.
Article in English | MEDLINE | ID: mdl-31266934

ABSTRACT

BACKGROUND Subclinical epileptiform discharges (SEDs) are defined as epileptiform electroencephalographic (EEG) discharges without clinical signs of seizure in patients. The subthreshold convulsant discharge (SCD) is a frequently used model for SEDs. This study aimed to investigate the effect of levetiracetam (LEV), an anti-convulsant drug, on cognitive impairment of SCD model rats and to assess the associated mechanisms. MATERIAL AND METHODS A SCD rat model was established. Rats were divided into an SCD group, an SCD+ sodium valproate (VPA) group, and an SCD+ levetiracetam (LEV) group. The Morris water maze was used to evaluate the capacity of positioning navigation and space exploration. The field excitatory post-synaptic potentials (fEPSPs) were evaluated using a bipolar stimulation electrode. NCAM, GAP43, PS95, and CaMK II levels were detected using Western blot and RT-PCR, respectively. PKC activity was examined by a non-radioactive method. RESULTS LEV shortens the latency of platform seeking in SCD rats in positioning navigation. fEPSP slopes were significantly lower in the SCD group, and LEV treatment significantly enhanced the fEPSP slopes compared to the SCD group (P<0.05). The NCAM and GAP-43 levels were increased and PSD-95 levels were increased in SCD rats (P<0.05), which were improved by LEV treatment. The PKC activity and CaMK II levels were decreased in SCD rats and LEV treatment significantly enhanced PKC activity and increased CaMK II levels. CONCLUSIONS Cognitive impairment in of SCD model rats may be caused by decreased PKC activity, low expression of CaMK II, and inhibition of LTP formation. LEV can improve cognitive function by activating the PKC-GAP-43-CaMK signal transduction pathway.


Subject(s)
Cognitive Dysfunction/metabolism , Cognitive Dysfunction/prevention & control , Levetiracetam/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cognitive Dysfunction/drug therapy , Disease Models, Animal , Electroencephalography , GAP-43 Protein/drug effects , GAP-43 Protein/metabolism , Hippocampus/metabolism , Levetiracetam/metabolism , Male , Phosphorylation , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Seizures/drug therapy , Signal Transduction/drug effects , Valproic Acid/therapeutic use
2.
Curr Opin Cell Biol ; 7(6): 798-805, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8608010

ABSTRACT

Evolutionarily conserved from yeast to man, mitogen-activated protein kinase (MAPK) pathways respond to a variety of disparate signals which induce differentiation, proliferation, or changes in intracellular enzyme regulation. Recent advances have identified two new mammalian MAPK relatives, JNK1 and p38, and the pathways which are responsible for their activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Heat-Shock Proteins/metabolism , Mitogens/pharmacology , Phosphoric Monoester Hydrolases/metabolism , Yeasts/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Heat-Shock Proteins/drug effects , Mammals , Yeasts/cytology , Yeasts/drug effects
3.
Nat Med ; 9(7): 914-20, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12819778

ABSTRACT

Streptococcus pyogenes-induced acute rheumatic fever (ARF) is one of the best examples of postinfectious autoimmunity due to molecular mimicry between host and pathogen. Sydenham chorea is the major neurological manifestation of ARF but its pathogenesis has remained elusive, with no candidate autoantigen or mechanism of pathogenesis described. Chorea monoclonal antibodies showed specificity for mammalian lysoganglioside and N-acetyl-beta-D-glucosamine (GlcNAc), the dominant epitope of the group A streptococcal (GAS) carbohydrate. Chorea antibodies targeted the surface of human neuronal cells, with specific induction of calcium/calmodulin-dependent protein (CaM) kinase II activity by monoclonal antibody 24.3.1 and sera from active chorea. Convalescent sera and sera from other streptococcal diseases in the absence of chorea did not activate the kinase. The new evidence implicates antibody-mediated neuronal cell signaling in the immunopathogenesis of Sydenham chorea and will lead to a better understanding of other antibody-mediated neurological disorders.


Subject(s)
Autoantibodies/metabolism , Chorea/immunology , G(M1) Ganglioside/analogs & derivatives , Molecular Mimicry , Neurons/metabolism , Signal Transduction/immunology , Acetylglucosamine/immunology , Adolescent , Antibodies, Monoclonal/pharmacology , Autoantibodies/immunology , Brain/immunology , Brain/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Chorea/metabolism , Enzyme Activation/drug effects , Female , G(M1) Ganglioside/immunology , Humans , Immune Sera , Neurons/immunology , Streptococcus/immunology
4.
J Cell Biol ; 168(6): 887-97, 2005 Mar 14.
Article in English | MEDLINE | ID: mdl-15767461

ABSTRACT

Class II histone deacetylases (HDACs) may decrease slow muscle fiber gene expression by repressing myogenic transcription factor myocyte enhancer factor 2 (MEF2). Here, we show that repetitive slow fiber type electrical stimulation, but not fast fiber type stimulation, caused HDAC4-GFP, but not HDAC5-GFP, to translocate from the nucleus to the cytoplasm in cultured adult skeletal muscle fibers. HDAC4-GFP translocation was blocked by calmodulin-dependent protein kinase (CaMK) inhibitor KN-62. Slow fiber type stimulation increased MEF2 transcriptional activity, nuclear Ca(2+) concentration, and nuclear levels of activated CaMKII, but not total nuclear CaMKII or CaM-YFP. Thus, calcium transients for slow, but not fast, fiber stimulation patterns appear to provide sufficient Ca(2+)-dependent activation of nuclear CaMKII to result in net nuclear efflux of HDAC4. Nucleocytoplasmic shuttling of HDAC4-GFP in unstimulated resting fibers was not altered by KN-62, but was blocked by staurosporine, indicating that different kinases underlie nuclear efflux of HDAC4 in resting and stimulated muscle fibers.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation, Enzymologic , Histone Deacetylases/metabolism , Muscle, Skeletal/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Active Transport, Cell Nucleus , Adenoviridae/genetics , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cell Nucleus/enzymology , Cells, Cultured , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electric Stimulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Fluorescence Recovery After Photobleaching , Green Fluorescent Proteins/metabolism , Histone Deacetylases/classification , Histone Deacetylases/genetics , Kinetics , MEF2 Transcription Factors , Mice , Mice, Inbred Strains , Microscopy, Confocal , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/cytology , Myogenic Regulatory Factors , Recombinant Fusion Proteins/metabolism , Staurosporine/pharmacology , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Acta Pharmacol Sin ; 31(1): 93-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20048748

ABSTRACT

AIM: To investigate the apoptosis-inducing effect of trichostatin A (TSA) in the human lung adenocarcinoma cisplatin-resistant cell line (A549/CDDP) and to examine whether TSA can enhance sensitivity to cisplatin treatment and the underlying molecular mechanisms of such an enhancement. METHODS: Cell viability was evaluated using the Neutral Red assay. Apoptosis was assessed using Hoechst 33258 staining and flow cytometry analysis. Protein expression was detected by Western blotting. To determine the role of Death-associated protein kinase (DAPK) in TSA-induced apoptosis in the A549/CDDP cell line, cells were transfected with pcDNA3.1(+)-DAPK, which has a higher expression level of DAPK compared to endogenous expression, and DAPK activity was inhibited by both over-expression C-terminal fragment of DAPK which may competitive binding DAPK substrates to inhibit the function of DAPK and RNA interference. RESULTS: TSA induced apoptosis in both A549 cells and A549/CDDP cells. TSA enhanced the sensitivity of A549/CDDP cells to cisplatin, along with concomitant DAPK up-regulation. When DAPK was over-expressed, A549/CDDP cells became sensitive to cisplatin and the cytotoxicity of TSA could be increased. Moreover, the cytotoxicity of TSA could be alleviated by inhibition of DAPK activity by the expression of a recombinant C-terminal fragment of DAPK or RNA interference. CONCLUSION: TSA induced sensitivity to cisplatin treatment in cisplatin-resistant A549 cells. The up-regulation of DAPK is one of the mechanisms mediating sensitization to TSA-induced apoptosis in cisplatin-resistant cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Hydroxamic Acids/pharmacology , Protein Synthesis Inhibitors/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/physiopathology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line, Tumor , Cell Survival/drug effects , Death-Associated Protein Kinases , Drug Resistance, Neoplasm/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/physiopathology , Up-Regulation/drug effects
6.
Neuron ; 45(6): 837-45, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15797546

ABSTRACT

Developmental axon competition plays a key role in sculpting neural circuitry. Here, we have asked how activity and neurotrophins could interact to select one axon over another. Using compartmented cultures of sympathetic neurons, we show that, in the presence of NGF, local depolarization confers a competitive growth advantage on the depolarized axon collaterals and at the same time disadvantages the growth of unstimulated axons from the same and competing neurons. Depolarization mediates the competitive advantage by activating a CaMKII-MEK pathway, which converges to enhance local NGF-mediated downstream growth signals. Patterned electrical stimulation also acts via this pathway to enhance NGF-promoted axonal growth. In contrast, the competitive disadvantage is due to BDNF secreted from and acting on the unstimulated, competing axons through p75NTR. Thus, activity regulates both positive and negative neurotrophin-derived signaling cascades to confer a competitive growth advantage on one axon versus another, thereby providing a cellular mechanism for developmental axon selection.


Subject(s)
Action Potentials/physiology , Cell Differentiation/physiology , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Neural Pathways/growth & development , Superior Cervical Ganglion/growth & development , Animals , Animals, Newborn , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Communication/physiology , Cell Differentiation/drug effects , Cells, Cultured , Growth Cones/drug effects , MAP Kinase Kinase 1/drug effects , MAP Kinase Kinase 1/metabolism , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Nerve Growth Factors/pharmacology , Neural Pathways/cytology , Neural Pathways/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/metabolism
7.
Trends Biochem Sci ; 19(11): 470-3, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7855889

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are activated by dual phosphorylation on threonine and tyrosine in response to a wide array of extracellular stimuli. In the yeast Saccharomyces cerevisiae, a series of extracellular stimuli. In the yeast Saccharomyces cerevisiae, a series of MAPK signal transduction pathways have been demonstrated to control many cellular functions. By contrast, mammalian MAPKs are more poorly understood. However, recent studies have established important roles for three separate groups of mammalian MAPKs, which are characterized by distinct dual phosphorylation motifs. Together, these protein kinases mediate signal transduction in mammalian tissues and control many aspects of cellular physiology.


Subject(s)
Mitogen-Activated Protein Kinases , Mitogens/pharmacology , Protein Kinases/drug effects , Amino Acid Sequence , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Molecular Sequence Data
8.
Br J Pharmacol ; 154(4): 729-40, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18454172

ABSTRACT

Multifunctional Ca2+/calmodulin-dependent protein kinases (CaMKs) play pivotal roles in intracellular Ca2+ signaling pathways. There is growing evidence that CaMKs are involved in the pathogenic mechanisms underlying various human diseases. In this review, we begin by briefly summarizing our knowledge of the involvement of CaMKs in the pathogenesis of various diseases suggested to be caused by the dysfunction/dysregulation or aberrant expression of CaMKs. It is widely known that the activities of CaMKs are strictly regulated by protein phosphorylation/dephosphorylation of specific phosphorylation sites. Since phosphorylation status is balanced by protein kinases and protein phosphatases, the mechanism of dephosphorylation/deactivation of CaMKs, corresponding to their 'switching off', is extremely important, as is the mechanism of phosphorylation/activation corresponding to their 'switching on'. Therefore, we focus on the regulation of multifunctional CaMKs by protein phosphatases. We summarize the current understanding of negative regulation of CaMKs by protein phosphatases. We also discuss the biochemical properties and physiological significance of a protein phosphatase that we designated as Ca2+/calmodulin-dependent protein kinase phosphatase (CaMKP), and those of its homologue CaMKP-N. Pharmacological applications of CaMKP inhibitors are also discussed. These compounds may be useful not only for exploring the physiological functions of CaMKP/CaMKP-N, but also as novel chemotherapies for various diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Phosphoprotein Phosphatases/pharmacology , Signal Transduction/drug effects , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Humans , Phosphoprotein Phosphatases/metabolism , Phosphorylation
9.
Nat Neurosci ; 3(9): 881-6, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966618

ABSTRACT

Synaptic plasticity is thought to be a key process for learning, memory and other cognitive functions of the nervous system. The initial events of plasticity require the conversion of brief electrical signals into alterations of the biochemical properties of synapses that last for much longer than the initial stimuli. Here we show that a regulator of synaptic plasticity, calcium/calmodulin-dependent protein kinase IIalpha (CaMKII), sequentially translocates to postsynaptic sites, undergoes autophosphorylation and gets trapped for several minutes until its dissociation is induced by secondary autophosphorylation and phosphatase 1 action. Once dissociated, CaMKII shows facilitated translocation for several minutes. This suggests that trapping of CaMKII by its targets and priming of CaMKII translocation may function as biochemical memory mechanisms that change the signaling capacity of synapses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Memory/physiology , Neuronal Plasticity/physiology , Synaptic Membranes/metabolism , Aniline Compounds , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cells, Cultured , Electric Stimulation , Fluorescent Dyes , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Green Fluorescent Proteins , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/ultrastructure , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Phosphorylation , Synaptic Membranes/drug effects , Synaptic Membranes/ultrastructure , Xanthenes
10.
Curr Biol ; 7(1): 63-70, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-8999998

ABSTRACT

BACKGROUND: The small GTPase R-Ras displays a less potent transforming activity than the closely related Ras oncogene products. Although R-Ras has been reported to interact with c-Raf1 and Ral-GDS in vitro, the pathways by which it exerts its effects on cellular proliferation are not known. RESULTS: Both Ras and R-Ras interact with phosphoinositide (PI) 3-kinase in vitro, and induce elevation of the levels of PI 3-kinase lipid products in intact cells. Unlike Ras, R-Ras does not activate Raf or mitogen-activated protein (MAP) kinase in cells. In co-transfection assays, the serine/threonine protein kinase PKB (or Akt) is effectively stimulated by R-Ras, Ras, mutants of Ras that activate PI 3-kinase but not other effectors, and activated forms of PI 3-kinase. Ras and R-Ras stimulate PKB/Akt through a non-autocrine mechanism that involves PI 3-kinase. The constitutive activation of PI 3-kinase alone is sufficient to activate PKB/Akt, but not the MAP kinase ERK or the stress-activated protein kinase, Jun N-terminal kinase. Transformation assays in fibroblasts suggest that PKB/Akt and Raf are part of distinct oncogenic signalling pathways. CONCLUSIONS: Both the Raf-MAP kinase and PI 3-kinase-PKB/Akt pathways are activated by Ras, but only the PI 3-kinase-PKB/Akt pathway is activated by R-Ras. PI 3-kinase, and downstream targets such as PKB/Akt, are likely to be essential mediators of transformation induced by R-Ras. PI 3-kinase, as well as Raf, is thus implicated also in Ras transformation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/drug effects , GTP Phosphohydrolases/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/drug effects , ras Proteins/pharmacology , Animals , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic/physiology , Mice , Phosphatidylinositol 3-Kinases
11.
J Clin Invest ; 98(8): 1897-905, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8878442

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation and migration are responses to arterial injury that are highly important to the processes of restenosis and atherosclerosis. In the arterial balloon injury model in the rat, platelet-derived growth factor (PDGF) and basic fibroblast growth factor (bFGF) are induced in the vessel wall and regulate these VSMC activities. Novel insulin sensitizing agents, thiazolidinediones, have been demonstrated to inhibit insulin and epidermal growth factor-induced growth of VSMCs. We hypothesized that these agents might also inhibit the effect of PDGF and bFGF on cultured VSMCs and intimal hyperplasia in vivo. Troglitazone (1 microM), a member of the thiazolidinedione class, produced a near complete inhibition of both bFGF-induced DNA synthesis as measured by bromodeoxyuridine incorporation (6.5+/-3.9 vs. 17.6+/-4.3% cells labeled, P < 0.05) and c-fos induction. This effect was associated with an inhibition (by 73+/-4%, P < 0.01) by troglitazone of the transactivation of the serum response element, which regulates c-fos expression. Inhibition of c-fos induction by troglitazone appeared to occur via a blockade of the MAP kinase pathway at a point downstream of MAP kinase activation by MAP kinase kinase. At this dose, troglitazone also inhibited PDGF-BB-directed migration of VSMC (by 70+/-6%, P < 0.01). These in vitro effects were operative in vivo. Quantitative image analysis revealed that troglitazone-treated rats had 62% (P < 0.001) less neointima/media area ratio 14 d after balloon injury of the aorta compared with injured rats that received no troglitazone. These results suggest troglitazone is a potent inhibitor of VSMC proliferation and migration and, thus, may be a useful agent to prevent restenosis and possibly atherosclerosis.


Subject(s)
Chromans/pharmacology , Hypoglycemic Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Thiazoles/pharmacology , Thiazolidinediones , Animals , Arteriosclerosis/prevention & control , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , DNA/biosynthesis , Fibroblast Growth Factor 2/antagonists & inhibitors , Genes, fos/drug effects , Hyperplasia , Male , Muscle, Smooth, Vascular/pathology , Platelet-Derived Growth Factor/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Troglitazone
12.
Psychopharmacology (Berl) ; 191(2): 323-31, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17160679

ABSTRACT

RATIONALE: Females have been demonstrated repeatedly to be more sensitive to cocaine. The role of the frontal cortex (FCX) in mediating behavioral sensitization and the underlying signaling pathways are unclear. OBJECTIVE: The study was designed to characterize the role of FCX calcium/calmodulin-dependent protein kinase II (CaMKII) activity in the behavioral supersensitization observed in female rats after prolonged cocaine exposure. MATERIALS AND METHODS: Intact female rats that received cocaine for 9 days followed by 7 days of drug withdrawal constituted the model used for studying the mechanism of supersensitization. RESULTS: This cocaine withdrawal treatment resulted in behavioral supersensitization in intact female rats as indicated by an enhanced behavioral response to cocaine challenge assessed on day 16 (7-day withdrawal) and compared to the response on day 9 of cocaine treatment. This treatment regimen did not lead to supersensitization in male or in ovariectomized (OVX) rats. Administration of estrogen to OVX rats restored behavioral supersensitivity to repeated cocaine. FCX CaMKII activity was significantly altered by cocaine in females, and this effect was related to estrogen's presence; cocaine-induced changes in striatal CaMKII activity were, however, less estrogen-sensitive. Furthermore, estrogen-modulated FCX CaMKII activity in cocaine-supersensitized rats was dependent on D(1) dopamine receptor activation. CONCLUSION: Estrogen-modulated D(1) dopamine receptor activity mediates the effects of prolonged cocaine exposure on FCX CaMKII, and this, in turn, may contribute to the development of behavioral supersensitivity to repeated cocaine treatment in intact female rats.


Subject(s)
Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Estradiol/analogs & derivatives , Receptors, Dopamine D1/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Drug Administration Schedule , Estradiol/pharmacology , Estrogens/pharmacology , Female , Male , Ovariectomy , Prefrontal Cortex , Pyramidal Cells , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism , Sex Factors , Signal Transduction/drug effects
13.
Neurosci Lett ; 423(2): 143-8, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17669591

ABSTRACT

Ca(2+)/calmodulin-dependent protein kinase kinases (CaMKKs) are upstream protein kinases that phosphorylate and activate CaMKI and CaMKIV, both of which are involved in a variety of neuronal functions. Here, we first demonstrated that the two isoforms of CaMKK were differentially expressed during neural development by in situ hybridization. We also demonstrated that both dominant negative and pharmacological interference with CaMKK inhibitor, STO-609 resulted in a significant decrease in the number of primary dendrites of cultured hippocampal neurons. Our present findings provide the detailed anatomical information on the developmental expression of CaMKKs and the functional involvement of CaMKK in the formation of primary dendrites.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Dendrites/enzymology , Hippocampus/embryology , Hippocampus/enzymology , Animals , Benzimidazoles/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cells, Cultured , Dendrites/drug effects , Embryo, Mammalian , In Situ Hybridization , Isoenzymes/biosynthesis , Isoquinolines/pharmacology , Mice , Microscopy, Confocal , Naphthalimides/pharmacology , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
14.
Mol Cell Biol ; 22(13): 4522-34, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12052862

ABSTRACT

Retinoids exhibit antineoplastic activities that may be linked to retinoid receptor-mediated transrepression of activating protein 1 (AP1), a heterodimeric transcription factor composed of fos- and jun-related proteins. Here we show that transcriptional activation of an AP1-regulated gene through the mitogen-activated protein kinase (MAPK)-extracellular signal-regulated kinase (ERK) pathway (MAPK(ERK)) is characterized, in intact cells, by a switch from a fra2-junD dimer to a junD-fosB dimer loading on its promoter and by simultaneous recruitment of ERKs, CREB-binding protein (CBP), and RNA polymerase II. All-trans-retinoic acid (atRA) receptor (RAR) was tethered constitutively to the AP1 promoter. AP1 transrepression by retinoic acid was concomitant to glycogen synthase kinase 3 activation, negative regulation of junD hyperphosphorylation, and to decreased RNA polymerase II recruitment. Under these conditions, fra1 loading to the AP1 response element was strongly increased. Importantly, CBP and ERKs were excluded from the promoter in the presence of atRA. AP1 transrepression by retinoids was RAR and ligand dependent, but none of the functions required for RAR-mediated transactivation was necessary for AP1 transrepression. These results indicate that transrepressive effects of retinoids are mediated through a mechanism unrelated to transcriptional activation, involving the RAR-dependent control of transcription factors and cofactor assembly on AP1-regulated promoters.


Subject(s)
MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Retinoic Acid/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Anisomycin/pharmacology , CREB-Binding Protein , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA/metabolism , Dimerization , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , HeLa Cells/drug effects , HeLa Cells/radiation effects , Humans , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/radiation effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Response Elements , Retinoic Acid Receptor alpha , Retinoid X Receptors , Tetradecanoylphorbol Acetate/pharmacology , Trans-Activators/genetics , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/genetics , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology , Ultraviolet Rays
15.
Peptides ; 27(11): 2738-49, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16914228

ABSTRACT

Leptin, an adipocytokine encoded by an obesity gene and expressed in adipose tissue, affects feeding behavior, thermogenesis, and neuroendocrine status via leptin receptors distributed in the brain, especially in the hypothalamus. Leptin may also modulate the synaptic plasticity and behavioral performance related to learning and memory since: leptin receptors are found in the hippocampus, and both leptin and its receptor share structural and functional similarities with the interleukin-6 family of cytokines that modulate long-term potentiation (LTP) in the hippocampus. We therefore examined the effect of leptin on (1) behavioral performance in emotional and spatial learning tasks, (2) LTP at Schaffer collateral-CA1 synapses, (3) presynaptic and postsynaptic activities in hippocampal CA1 neurons, (4) the intracellular Ca(2+) concentration ([Ca(2+)](i)) in CA1 neurons, and (5) the activity of Ca(2+)/calmodulin protein kinase II (CaMK II) in the hippocampal CA1 tissue that exhibits LTP. Intravenous injection of 5 and/or 50mug/kg, but not of 500mug/kg leptin, facilitated behavioral performance in passive avoidance and Morris water-maze tasks. Bath application of 10(-12)M leptin in slice experiments enhanced LTP and increased the presynaptic transmitter release, whereas 10(-10)M leptin suppressed LTP and reduced the postsynaptic receptor sensitivity to N-methyl-d-aspartic acid. The increase in the [Ca(2+)](i) induced by 10(-10)M leptin was two times greater than that induced by 10(-12)M leptin. In addition, the facilitation (10(-12)M) and suppression (10(-10)M) of LTP by leptin was closely associated with an increase and decrease in Ca(2+)-independent activity of CaMK II. Our results show that leptin not only affects hypothalamic functions (such as feeding, thermogenesis, and neuroendocrine status), but also modulates higher nervous functions, such as the behavioral performance related to learning and memory and hippocampal synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Hippocampus/physiology , Leptin/pharmacology , Long-Term Potentiation/physiology , Maze Learning/drug effects , Memory/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Hippocampus/drug effects , Leptin/physiology , Long-Term Potentiation/drug effects , Male , Neurons/drug effects , Neurons/physiology , Phosphorylation/drug effects , Rats , Rats, Wistar
16.
Brain Res ; 1108(1): 98-106, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16843447

ABSTRACT

Lithium used in bipolar mood disorder therapy protects neurons from brain ischemic cell death. Here, we documented that lithium administration under microsphere-embolism (ME)-induced brain ischemia restored decreased protein kinase B (Akt) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) activities 24 h after ischemia in rat brain. Akt activation was associated with increased phosphorylation of its potential targets forkhead transcription factor (FKHR) and glycogen synthase kinase-3beta (GSK-3beta). In parallel with decreased CaMKII autophosphorylation, we also found marked dephosphorylation of tau proteins 24-72 h after ME. Increased protein phosphatase 2A (PP2A) activity was found 24 h after ME. Inhibition of increased PP2A activity by lithium treatment apparently mediated restored tau phosphorylation. Taken together, activation of Akt and CaMKII by lithium was associated with neuroprotective activity in ME-induced neuronal injury.


Subject(s)
Brain Infarction/drug therapy , Brain Ischemia/drug therapy , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Lithium Chloride/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Animals , Antimanic Agents/pharmacology , Brain Infarction/metabolism , Brain Infarction/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/metabolism , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Intracranial Embolism/drug therapy , Intracranial Embolism/metabolism , Intracranial Embolism/physiopathology , Male , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Phosphoprotein Phosphatases/drug effects , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2 , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Serine/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , tau Proteins/drug effects , tau Proteins/metabolism
17.
Circ Res ; 88(5): 513-9, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11249875

ABSTRACT

Glucose-insulin-potassium solutions exert beneficial effects on the ischemic heart by reducing infarct size and mortality and improving postischemic left ventricular function. Insulin could be the critical protective component of this mixture, although the insulin response of the ischemic and postischemic myocardium has not been systematically investigated. The aim of this work was to study the insulin response during ischemia by analyzing insulin signaling. This was evaluated by measuring changes in activity and/or phosphorylation state of insulin signaling elements in isolated perfused rat hearts submitted to no-flow ischemia. Intracellular pH (pH(i)) was measured by NMR. No-flow ischemia antagonized insulin signaling including insulin receptor, insulin receptor substrate-1, phosphatidylinositol 3-kinase, protein kinase B, p70 ribosomal S6 kinase, and glycogen synthase kinase-3. These changes were concomitant with intracellular acidosis. Perfusing hearts with ouabain and amiloride in normoxic conditions decreased pH(i) and insulin signaling, whereas perfusing at pH 8.2 counteracted the drop in pH(i) and the inhibition of insulin signaling by ischemia. Incubation of cardiomyocytes in normoxic conditions, but at pH values below 6.75, mimicked the effect of ischemia and also inhibited insulin-stimulated glucose uptake. Finally, the in vitro insulin receptor tyrosine kinase activity was progressively inhibited at pH values below physiological pH(i), being abolished at pH 6.0. Therefore, ischemic acidosis decreases kinase activity and tyrosine phosphorylation of the insulin receptor thereby preventing activation of the downstream components of the signaling pathway. We conclude that severe ischemia inhibits insulin signaling by decreasing pH(i).


Subject(s)
Heart/drug effects , Insulin/pharmacology , Myocardial Ischemia/physiopathology , Protein Serine-Threonine Kinases , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Heart/physiology , Hydrogen-Ion Concentration , Insulin Receptor Substrate Proteins , Male , Myocardial Reperfusion , Myocardium/cytology , Myocardium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Receptor, Insulin/metabolism , Ribosomal Protein S6 Kinases/drug effects , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction
18.
Circ Res ; 88(1): 88-96, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11139479

ABSTRACT

Mitogen-activated protein kinase (MAPK) signaling pathways are important regulators of cell growth, proliferation, and stress responsiveness. A family of dual-specificity MAP kinase phosphatases (MKPs) act as critical counteracting factors that directly regulate the magnitude and duration of p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) activation. Here we show that constitutive expression of MKP-1 in cultured primary cardiomyocytes using adenovirus-mediated gene transfer blocked the activation of p38, JNK1/2, and ERK1/2 and prevented agonist-induced hypertrophy. Transgenic mice expressing physiological levels of MKP-1 in the heart showed (1) no activation of p38, JNK1/2, or ERK1/2; (2) diminished developmental myocardial growth; and (3) attenuated hypertrophy in response to aortic banding and catecholamine infusion. These results provide further evidence implicating MAPK signaling factors as obligate regulators of cardiac growth and hypertrophy and demonstrate the importance of dual-specificity phosphatases as counterbalancing regulatory factors in the heart.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cardiomegaly/enzymology , Myocardium/enzymology , Protein Tyrosine Phosphatases/metabolism , Adenoviridae/genetics , Animals , Animals, Newborn , Atrial Natriuretic Factor/drug effects , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Catecholamines/pharmacology , Cells, Cultured , DNA, Recombinant , Endothelin-1/pharmacology , Female , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Mice , Mice, Transgenic , Myocardium/cytology , Myocardium/metabolism , Phenylephrine/pharmacology , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/genetics , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
19.
Cancer Res ; 54(1): 12-5, 1994 Jan 01.
Article in English | MEDLINE | ID: mdl-8261431

ABSTRACT

Extracellular signal-regulated kinases (ERKs), also known as mitogen-activated protein (MAP) kinases, are rapidly phosphorylated and activated in response to a number of external factors which promote growth and differentiation (T. G. Boulton, S. H. Nye, D. J. Robbins, N. Y. Ip, E. Radziejewska, S. D. Morgenbesser, R. A. DePinho, N. Panayotatos, M. H. Cobb, and G. D. Yancopoulos, Cell, 65: 663-675, 1991; S. L. Pelech and S. S. Jasbinder, Science (Washington DC), 257: 1355-1356, 1992; G. Thomas, Cell, 68: 3-6, 1992). We have identified two novel stimulators of MAP kinase activity, ionizing radiation and H2O2. Both radiation and H2O2, as well as the known agonist 12-O-tetradecanoylphorbol 13-acetate activate MAP kinase through the production of reactive oxygen intermediates. Our results demonstrate a direct link between the MAP kinase signal transduction pathway and reactive oxygen species and provide a unifying mechanism for activation of early- and late-response genes by inducers of oxidative stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , 3T3 Cells , Acetylcysteine/pharmacology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/radiation effects , Down-Regulation , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Hydrogen Peroxide/pharmacology , Mice , Oxidation-Reduction , Phosphorylation , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology
20.
Cancer Res ; 57(21): 4714-7, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9354428

ABSTRACT

The activation of c-Jun NH2-terminal kinase (JNK)/stress-activated protein kinase (SAPK) and/or extracellular signal-regulated kinase (ERK) is involved in ceramide-induced apoptosis in certain cells. To examine the relationship between activated Ki-ras-mediated signals and ceramide-induced apoptosis in human colon cancer cells, JNK/SAPK and ERK activity, as initiated by ceramide, was examined in HCT116, which has a mutation of Ki-ras at codon 13, and HCT116-derived clones, HKe-3 and HKh-2, in which activated Ki-ras was disrupted through gene targeting. In HKe-3 and HKh-2, the activity of JNK/SAPK increased significantly within 60 min following C2 ceramide stimulation, and some apoptosis followed. In contrast, C2 ceramide caused a marked apoptosis in HCT116, but activation of JNK/SAPK was not observed. C2 ceramide did not activate ERK in any of the cell lines. These results suggest that activated Ki-ras contributes to the sensitivity of ceramide-induced apoptosis without JNK/SAPK or ERK activation and that other signaling pathways involved in ceramide-induced apoptosis may be present in human colon cancer cells.


Subject(s)
Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Ceramides/pharmacology , Colonic Neoplasms/enzymology , Genes, ras/physiology , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-raf/metabolism , Apoptosis/genetics , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Colonic Neoplasms/pathology , DNA Fragmentation , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , JNK Mitogen-Activated Protein Kinases , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL