Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Inhal Toxicol ; 24(11): 741-50, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22954398

ABSTRACT

Fly ash from industrial waste incinerators has been a significant concern because of their constituent toxic heavy metals and organic compounds. The objective of this study was to identify the subacute inhalation toxicity of fly ash from industrial waste incinerators, using whole body inhalation exposure chambers. Male and female groups of Sprague-Dawley rats were exposed to fly ash by inhalation of concentrations of 0, 50, 100, 200 mg/m(3), for 6 h/day, 5 days/week for 4 weeks. There was no significant difference in body weight, and relative organ weight to body weight, between the exposure groups and the control group. Hematological examinations revealed a significant increase of monocyte counts in fly ash exposed rats and brown pigment laden macrophage was found in the lungs of rats exposed to high concentration of fly ash. A decrease of blood glucose levels and an increase in glutamate oxaloacetate transaminase activity were observed in fly ash treated rats. There was also a significant increase of lactate dehydrogenase levels in rat blood exposed fly ash. A significant dose-dependent increase of DNA damage was found in lymphocytes, spleen, bronchoalveolar lavage, liver, lung, and thymus of rats exposed to fly ash. In addition, the level of lipid peroxidation was increased in the plasma of rats exposed to a high concentration of fly ash. These results suggest that inhalation of fly ash from industrial waste incinerators can induce histopathologic, hematological, and serum biochemical changes and oxidative damage.


Subject(s)
Air Pollutants/toxicity , Coal Ash/toxicity , Incineration , Industrial Waste/analysis , Animals , Female , Inhalation Exposure , Lipid Peroxidation , Male , Malondialdehyde/blood , Rats , Rats, Sprague-Dawley
2.
Genes Genomics ; 44(5): 571-582, 2022 05.
Article in English | MEDLINE | ID: mdl-35254656

ABSTRACT

BACKGROUND: Calcium ions play a pivotal role in cell proliferation, differentiation, and migration. Under basal conditions, the calcium level is tightly regulated; however, cellular activation by growth factors increase the ion level through calcium pumps in the plasma membrane and endoplasmic reticulum for calcium signaling. Orai1 is a major calcium channel in the cell membrane of non-excitable cells, and its activity depends on the stromal interaction molecule 1 (Stim1). Several groups reported that the store-operated calcium entry (SOCE) can be modulated through phosphorylation of Stim1 by protein kinases such as extracellular signal-regulated kinase (ERK), protein kinase A (PKA), and p21-activated kinase (PAK). PKC is a protein kinase that is activated by calcium and diacylglycerol (DAG), but it remains unclear what role activated PKC plays in controlling the intracellular calcium pool. OBJECTIVES: Here, we investigated whether PKC-ß controls intracellular calcium dynamics through Stim1. METHODS: Several biochemical methods such as immune-precipitation, site directed mutagenesis, in vitro kinase assay were employed to investigate PKC interaction with and phosphorylation of Stim1. Intracellular calcium mobilization, via Stim1 mediated SOCE channel, were studied using in the presence of PKC activator or inhibitor under a confocal microscope. RESULTS: Our data demonstrate that PKC interacts with and phosphorylates Stim1 in vitro. phosphorylation of Stim1 at its C-terminal end appears to be important in the regulation of SOCE activity in HEK293 and HeLa cells. Additionally, transient intracellular calcium mobilization assays demonstrate that the SOCE activity was inhibited by PKC activators or activated by PKC inhibitors. CONCLUSION: In sum, our data suggest a repressive role of PKC in regulating calcium entry through SOCE.


Subject(s)
Calcium , Neoplasm Proteins , Calcium/metabolism , HEK293 Cells , HeLa Cells , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
3.
J Cell Physiol ; 226(1): 194-204, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20658516

ABSTRACT

Induction of terminal differentiation represents a potentially less toxic cancer therapy. Treatment of HO-1 human metastatic melanoma cells with IFN-ß plus mezerein (MEZ) promotes terminal differentiation with an irreversible loss of growth potential. During this process, the transcription factor FOXM1 is down-regulated potentially inhibiting transactivation of target genes including those involved in G(2)/M progression and cell proliferation. We investigated the mechanism of FOXM1 down-regulation and its physiological role in terminal differentiation. Genetic and pharmacological studies revealed that FOXM1 down-regulation was primarily caused by MEZ activation of PKCα and co-treatment with IFN-ß plus MEZ augmented the effect of PKCα. Promoter analysis with a mutated E-box on the FOXM1 promoter, and in vitro and in vivo binding assays confirm a direct role of c-Myc on FOXM1 expression. Reduction of c-Myc and overexpression of Mad1 by IFN-ß plus MEZ treatment should cause potent and persistent reduction of FOXM1 expression during terminal differentiation. Overexpression of FOXM1 restored expression of cell cycle-associated genes and increased the proportion of cells in the S phase. Our experiments support a model for terminal differentiation in which FOXM1 down-regulation via activation of PKCα followed by suppression of c-Myc expression, are causal events in promoting growth inhibition during terminal differentiation.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Melanoma/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Diterpenes/pharmacology , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Humans , Interferon-beta/pharmacology , Melanoma/pathology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction
4.
Mol Cell Biochem ; 351(1-2): 149-56, 2011 May.
Article in English | MEDLINE | ID: mdl-21240543

ABSTRACT

Protein kinases and protein phosphatases constitute about 2-4% of the genes in a typical eukaryotic genome. Protein phosphatases are important players in many cellular processes such as proliferation, differentiation, cell adhesion, and motility. In this study, we identified, classified, and analyzed protein phosphatase complement of the dog genome. In this article, we report the identification of at least 178 putative protein phosphatases in dog which include 51 PSTPs, 112 PTPs, and 15 Asp-based protein phosphatases. Interestingly, we found at least five novel protein phosphatases in dog, namely DUSP5L, DUSP18L, MTMR9L, MTMR12L, and PPP6CL which are not present in human, mouse, rat, and cow. In addition, we found PTP4A1-rt, a retro-transposed copy of the PTP4A1 gene, in chromosome 27. Furthermore, we modeled three-dimensional structures of the catalytic domains of these putative protein phosphatases and aligned them to see the structural similarities between them. We docked PPP2CA with okadaic acid and calculated the value of affinity energy as -8.8 kcal/mol. Our nucleotide substitution rate study revealed that apparently none of the phosphatase family is under significantly higher evolutionary pressure.


Subject(s)
Computational Biology , Genome , Phosphoprotein Phosphatases/genetics , Animals , Dogs , Models, Molecular , Open Reading Frames , Phosphoprotein Phosphatases/classification , Phosphoprotein Phosphatases/metabolism , Phylogeny , Subcellular Fractions/enzymology
5.
Arch Biochem Biophys ; 493(2): 242-8, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19914197

ABSTRACT

Protein kinase C delta (PKCdelta) is one of the important isoforms of PKCs that regulate various cellular processes, including cell survival and apoptosis. Studies have shown that activation of PKCdelta is correlated with apoptosis in various cell types, depending upon various stimuli. Phosphorylation of Thr505, Ser643 and Ser662 is crucial in activation of PKCdelta. Furthermore, phosphorylation of tyrosine residues, in particular that of Tyr311, is associated with PKCdelta activation and induction of apoptosis. Here, we generated a hydrophobic motif phosphorylation-deficient mutant of PKCdelta (PKCdelta-S662A) by mutating Ser662 to Ala, and studied the effect of this mutation in inducing apoptosis in L929 murine fibroblasts. We report that this mutation renders PKCdelta apoptotically more active. Furthermore, we found that the mutant PKCdelta-S662A is tyrosine-phosphorylated and translocated to the membrane faster than its wild-type counterpart.


Subject(s)
Apoptosis/physiology , Cell Membrane/enzymology , Fibroblasts/enzymology , Mutation, Missense , Protein Kinase C-delta/metabolism , Amino Acid Motifs/physiology , Amino Acid Substitution , Animals , Cell Line , Cell Membrane/genetics , Enzyme Activation/genetics , Fibroblasts/cytology , Hydrophobic and Hydrophilic Interactions , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Phosphorylation/genetics , Protein Kinase C-delta/genetics , Protein Transport/physiology
6.
J Neurochem ; 110(4): 1310-20, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19519660

ABSTRACT

Activation of V1 vasopressin (VP) receptors prevents serum deprivation-induced apoptosis in neuronal H32 cells, partially through mitogen-activated protein kinase (MAPK) mediated Bad phosphorylation. In this study, we investigated the role of protein kinases C (PKC) and B (PKB) mediating VP-induced antiapoptosis in H32 cells. Serum deprivation increased PKCdelta but not PKCalpha or PKCbeta activity, while VP increased PKCalpha and PKCbeta without affecting PKCdelta activity. Inhibition of PKCdelta prevented caspase 3 activation, indicating that PKCdelta mediates the pro-apoptotic actions of serum deprivation. Simultaneous inhibition of PKCalpha and beta and MAPK abolished VP-induced Bad phosphorylation, but it only partially prevented caspase 3 inhibition. Complete abolition of the protective effect of VP on serum deprivation-induced caspase 3 activity required additional blockade of phosphoinositide 3 kinase (PI3K)/protein kinase B. The data demonstrate that VP exerts antiapoptosis through multiple pathways; while PKCalpha and beta together with extracellular signal-regulated kinases/MAPK activation mediates Bad phosphorylation (inactivation), the full protective action of VP requires additional activation of PKB (PI3K/protein kinase B) pathway.


Subject(s)
Apoptosis/physiology , Cytoprotection/physiology , Neurons/metabolism , Neuroprotective Agents/metabolism , Protein Kinase C-alpha/metabolism , Vasopressins/metabolism , Animals , Apoptosis/drug effects , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Culture Media, Serum-Free/pharmacology , Cytoprotection/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Kinase C beta , Protein Kinase C-alpha/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Vasopressins/pharmacology , bcl-Associated Death Protein/drug effects , bcl-Associated Death Protein/metabolism
7.
Cancer Res ; 67(13): 6333-41, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616692

ABSTRACT

Heat shock protein 27 (HSP27), which is highly expressed in human lung and breast cancer tissues, induced resistance to cell death against various stimuli. Treatment of NCI-H1299 cells, which express a high level of HSP27, with small interference RNA specifically targeting HSP27 resulted in inhibition of their resistance to radiation or cisplatin, suggesting that HSP27 contributed to cellular resistance in these lung cancer cells. Furthermore, because HSP27 interacts directly with the COOH terminus of the protein kinase C delta (PKC delta)-V5 region with ensuing inhibition of PKC delta activity and PKC delta-mediated cell death, we wished to determine amino acid residues in the V5 region that mediate its interaction with HSP27. Investigation with various deletion mutants of the region revealed that amino acid residues 668 to 674 of the V5 region mediate its interaction with HSP27. When NCI-H1299 cells were treated with biotin or with FITC-tagged heptapeptide of the residues 668 to 674 (E-F-Q-F-L-D-I), the cells exhibited dramatically increased cisplatin or radiation-induced cell death with the heptapeptide having efficient interaction with HSP27, which in turn restored the PKC delta activity that had been inhibited by HSP27. In vivo nude mice grafting data also suggested that NCI-H1299 cells were sensitized by this heptapeptide. The above data strongly show that the heptapeptide of the PKC delta-V5 region sensitized human cancer cells through its interaction with HSP27, thereby sequestering HSP27. The heptapeptide may provide a novel strategy for selective neutralization of HSP27.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/metabolism , Neoplasm Proteins/metabolism , Oligopeptides/pharmacology , Protein Kinase C-delta/metabolism , Amino Acids/chemistry , Animals , Cell Line, Tumor , HSP27 Heat-Shock Proteins , Humans , Mice , Molecular Chaperones , Peptides/chemistry , Plasmids/metabolism , Transfection
8.
Cancers (Basel) ; 11(7)2019 Jul 11.
Article in English | MEDLINE | ID: mdl-31336725

ABSTRACT

Transmembrane Bax Inhibitor Motif-containing 6 (TMBIM6) is upregulated in several cancer types and involved in the metastasis. Specific downregulation of TMBIM6 results in cancer cell death. However, the TMBIM6 gene transcriptional regulation in normal and cancer cells is least studied. Here, we identified the core promoter region (-133/+30 bp) sufficient for promoter activity of TMBIM6 gene. Reporter gene expression with mutations at transcription factor binding sites, EMSA, supershift, and ChIP assays demonstrated that Sp1 is an essential transcription factor for basal promoter activity of TMBIM6. The TMBIM6 mRNA expression was increased with Sp1 levels in a concentration dependent manner. Ablation of Sp1 through siRNA or inhibition with mithramycin-A reduced the TMBIM6 mRNA expression. We also found that the protein kinase-C activation stimulates promoter activity and endogenous TMBIM6 mRNA by 2- to 2.5-fold. Additionally, overexpression of active mutants of PKCι, PKCε, and PKCδ increased TMBIM6 expression by enhancing nuclear translocation of Sp1. Immunohistochemistry analyses confirmed that the expression levels of PKCι, Sp1, and TMBIM6 were correlated with one another in samples from human breast, prostate, and liver cancer patients. Altogether, this study suggests the involvement of Sp1 in basal transcription and PKC in the enhanced expression of TMBIM6 in cancer.

9.
Cell Signal ; 53: 281-293, 2019 01.
Article in English | MEDLINE | ID: mdl-30352252

ABSTRACT

Conjugated linoleic acid (CLA) constitutes a group of isomers derived from linoleic acid. Diverse studies have suggested that these unsaturated fatty acids have beneficial effects on human health. However, it has also been reported that their consumption can generate alterations in hepatic tissue. Thus, in the present study, we evaluated the effect of two of the major isomers of CLA, cis-9, trans-11-CLA and trans-10, cis-12-CLA, in the regulation of insulin signaling in a hepatic cell model, clone 9 (C9). We found that the two isomers decrease insulin-stimulated phosphorylation of the main proteins involved in insulin signaling, such as Akt at Ser473 and Thr308, the insulin receptor at Tyr1158, IRS-1 at Tyr632, and GSK-3 at Ser9/21. Protein expression, however, was unaffected. Interestingly, both isomers of CLA promoted phosphorylation and activation of PKCε. Inhibition of PKCε activity by a dominant-negative form or knockdown of endogenous PKCε prevented the adverse effects of CLA isomers on insulin-induced Akt phosphorylation. Additionally, we also found that both isomers of CLA increase phosphorylation of IRS-1 at Ser612, a mechanism that probably underlies the inhibition of IRS-1 signaling by PKCε. Using confocal microscopy, we found that both isomers of CLA induced lipid accumulation in C9 cells with the presence of spherical cytosolic vesicles, suggesting their identity as neutral lipid droplets. These findings indicate that cis-9, trans-11-CLA and trans-10, cis-12-CLA isomers could have a significant role in the development of insulin resistance in hepatic C9 cells through IRS-1 serine phosphorylation, PKCε activation, and hepatic lipid accumulation.


Subject(s)
Insulin Resistance , Linoleic Acids, Conjugated/metabolism , Liver/cytology , Protein Kinase C-epsilon/metabolism , Animals , Cell Line , Enzyme Activation , Insulin/metabolism , Isomerism , Liver/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats
10.
Gene ; 410(1): 147-53, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18201844

ABSTRACT

Eukaryotic protein kinases, containing a conserved catalytic domain, represent one of the largest superfamilies of the eukaryotic proteins and play distinct roles in cell signaling and diseases. Near completion of rat genome sequencing project enables the evaluation of a near complete set of rat protein kinases. Publicly accessible genetic sequence databases were searched for rat protein kinases, and 515 eukaryotic protein kinases, 40 atypical protein kinases and 45 kinase pseudogenes were identified. The rat has 509 putative protein kinases orthologous to human kinases. Unlike microtubule affinity-regulating kinases, the rat has a few more kinases, in addition to the orthologous pairs of mouse kinases. The comparison of 11 different eukaryotic species revealed the evolutionary conservation of this diverse family of proteins. The evolutionary rate studies of human disease and non-disease associated kinases suggested that relatively uniform selective pressures have been applied to these kinase classes. This bioinformatic study of the rat protein kinases provides a suitable framework for further characterization of the functional and structural properties of these protein kinases.


Subject(s)
Computational Biology , Genome , Protein Kinases/metabolism , Animals , Humans , Open Reading Frames , Rats , Species Specificity
11.
Mol Carcinog ; 47(7): 519-25, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18163459

ABSTRACT

Although it is often assumed that the antitumor effects of nonsteroidal anti-inflammatory drugs (NSAIDs) are due to inhibition of cyclooxgenase (COX) activity, specifically COX-2, there is accumulating evidence that COX-2 independent mechanisms can also play an important role. Studies with sulindac sulfone (Aptosyn) and related derivatives have revealed a novel pathway of tumor growth inhibition and apoptosis mediated by activation of the guanosine 3',5' monophosphate (cGMP)-dependent enzyme protein kinase G (PKG). The present study indicates that concentrations of the NSAIDs celecoxib, indomethacin, and meclofenamic acid that inhibit growth of SW480 human colon cancer cells inhibit subcellular cGMP-phosphodiesterase (PDE) enzymatic activity and in intact cells induce a two- to threefold increase in intracellular levels of cGMP. This is associated with phosphorylation of the protein VASP, a marker of PKG activation, activation of JNK1 and a decrease in cellular levels of cyclin D1; effects seen with other agents that cause activation of PKG in these cells. On the other hand even a high concentration of the COX-2 specific inhibitor rofecoxib (500 microM) did not inhibit growth of SW480 cells. Nor did rofecoxib inhibit cGMP-PDE activity or cause other changes related to PKG activation in these cells. Since activation of the PKG pathways by celecoxib, indomethacin, and meclofenamic acid in this cell culture system required high concentrations of these compounds, it remains to be determined whether activation of this pathway contributes to the in vivo antitumor effects of specific NSAIDs.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/enzymology , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclooxygenase Inhibitors/pharmacology , Enzyme Activation/drug effects , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Celecoxib , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Cyclin D1/metabolism , Humans , Indomethacin/pharmacology , Lactones/pharmacology , Meclofenamic Acid/pharmacology , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Sulfones/pharmacology , Tumor Cells, Cultured
12.
Mol Cells ; 26(5): 462-7, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18719353

ABSTRACT

TPA is known to cooperate with an activated Ras oncogene in the transformation of rodent fibroblasts, but the biochemical mechanisms responsible for this effect have not been established. In the present study we used c-fos promoter-luciferase constructs as reporters, in transient transfection assays, in NIH3T3 cells to assess the mechanism of this cooperation. We found a marked synergistic interaction between TPA and a transfected v-Ha-ras oncogene in the activation of c-fos promoter and SRE. SRE has binding sites for TCF and SRF. A dominant-negative Ras (ras-N17) inhibited the TPA-Ras synergy by blocking the PKC-MAPK-TCF pathway. Dominant-negative RhoA and Rac1 (but not Cdc42Hs) inhibited the TPA-Ras synergy by blocking the Ras-Rho-SRF signaling pathway. Constitutively active PKCalpha and PKCepsilon showed synergy with v-Ras. These results suggest that the activation of two distinct pathways such as Ras-Raf-ERK-TCF pathway and Rho-SRF pathway are responsible for the induction of c-fos by TPA and Ras in mitogenic signaling pathways.


Subject(s)
Cell Nucleus/genetics , Genes, ras , Oncogene Protein p21(ras)/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , COS Cells , Cell Nucleus/drug effects , Chlorocebus aethiops , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Mice , NIH 3T3 Cells , Oncogene Protein p21(ras)/genetics , Protein Kinase C/metabolism , Serum Response Element , rho GTP-Binding Proteins/metabolism
13.
Endocrinology ; 148(3): 1099-107, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17185372

ABSTRACT

GnRH regulates the reproductive system by stimulating synthesis and release of gonadotropins. GnRH acts through a receptor coupled to multiple intracellular events including a rapid phosphoinositide turnover. Although the cAMP pathway is essential for gonadotrope function, the ability of GnRH to induce cAMP, as well as the coupling mechanisms involved, remain controversial. In this study, we established that GnRH increases intracellular cAMP levels in a concentration-dependent manner in LbetaT2 gonadotrope cells (maximal increase, 2.5-fold; EC(50), 0.30 nm), and this was further evidenced by GnRH activation of a cAMP-sensitive reporter gene. The GnRH effect was Ca(2+) independent, mimicked by the phorbol ester phorbol 12-myristate 13-acetate, and blocked by the protein kinase C (PKC) inhibitor bisindolylmaleimide, indicating that the GnRH effect was mediated by PKC. Pharmacological inhibition of conventional PKC isoforms with Gö6976 did not prevent GnRH-induced cAMP production, whereas down-regulation of novel PKCdelta, -epsilon, and -theta by a long-term treatment with GnRH markedly reduced it. Expression of dominant-negative (DN) mutants of PKCdelta or -epsilon but not PKCtheta impaired GnRH activation of a cAMP-sensitive promoter, demonstrating that PKCdelta and -epsilon are the two endogenous isoforms mediating GnRH activation of the adenylyl cyclase (AC) pathway in LbetaT2 cells. Accordingly, we identified by RT-PCR and immunocytochemical analysis, two PKC-sensitive AC isoforms, i.e. AC5 and AC7 as potential targets for GnRH. Lastly, we showed that only sustained stimulation of GnRH receptor significantly increased cAMP, suggesting that in vivo, the cAMP signaling pathway may be selectively recruited under intense GnRH release such as the preovulatory GnRH surge.


Subject(s)
Cyclic AMP/metabolism , Gonadotrophs , Gonadotropin-Releasing Hormone/physiology , Protein Kinase C-delta/physiology , Protein Kinase C-epsilon/physiology , Adenylyl Cyclases/metabolism , Animals , Calcium/physiology , Cell Line , Gonadotrophs/drug effects , Gonadotrophs/metabolism , Gonadotropin-Releasing Hormone/agonists , Isoenzymes/physiology , Mice , Protein Kinase C-delta/metabolism , Protein Kinase C-epsilon/metabolism , Receptors, LHRH/metabolism , Signal Transduction/drug effects
14.
Biochem Biophys Res Commun ; 364(2): 231-7, 2007 Dec 14.
Article in English | MEDLINE | ID: mdl-17942077

ABSTRACT

Protein kinase C (PKC), a multi-gene family of enzymes, plays key roles in the pathways of signal transduction, growth control and tumorigenesis. Variations in the intracellular localization of the individual isoforms are thought to be an important mechanism for the isoform-specific regulation of enzyme activity and substrate specificity. To provide a dynamic method of analyzing the localization of the specific isoforms of PKC in living cells, we generated fluorescent fusion proteins of the various PKC isoforms by using the green fluorescent protein (GFP) as a fluorescent marker at the carboxyl termini of these enzymes. The intracellular localization of the specific PKC isoforms was then examined by fluorescence microscopy after transient transfection of the respective PKC-GFP expression vector into NIH3T3 cells and subsequent TPA stimulation. We found that the specific isoforms of PKC display distinct localization patterns in untreated NIH3T3 cells. For example, PKCalpha is localized mainly in the cytoplasm while PKCepsilon is localized mainly in the Golgi apparatus. We also observed that PKCalpha, beta1, beta2, gamma, delta, epsilon, and eta translocate to the plasma membrane within 10 min of the start of TPA treatment, while the cellular localizations of PKCzeta and iota were not affected by TPA. Using a protein kinase inhibitor, we also showed that the kinase activity was not important for the translocation of PKC. These results suggest that specific PKC isoforms exert spatially distinct biological effects by virtue of their directed translocation to different intracellular sites.


Subject(s)
Protein Kinase C/physiology , Tetradecanoylphorbol Acetate/pharmacology , Animals , Cell Compartmentation , Cell Membrane/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Enzyme Activation , Golgi Apparatus/drug effects , Golgi Apparatus/physiology , Green Fluorescent Proteins/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/physiology , Mice , Microscopy, Fluorescence , NIH 3T3 Cells , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Staurosporine/pharmacology
15.
Oncogene ; 24(23): 3715-25, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15806174

ABSTRACT

Since radiation-induced caspase-dependent apoptosis and ROS generation were partially prevented by HSP25 overexpression, similar to the treatment of control cells with antioxidant agents such as DPI and tiron, questions arise whether radiation-mediated ROS generation contributes to the apoptotic cell death, and also whether HSP25 overexpression can reduce ROS mediated apoptotic cell death. In the present study, radiation-induced cytochrome c release from mitochondria and activation of caspases accompanied by a decrease of mitochondrial membrane potential in Jurkat T cells were shown to be inhibited by mitochondrial complex I inhibitor rotenone, suggesting that mitochondrial ROS might be important in radiation-induced caspase-dependent apoptosis. When HSP25 was overexpressed, effects similar to the treatment of cells with the antioxidants were obtained, indicating that HSP25 suppressed radiation-induced mitochondrial alteration that resulted in apoptosis. Furthermore, activation of p38 MAP kinase by radiation was associated with radiation-induced cell death and ROS production and PKCdelta was an upstream molecule for p38 MAP kinase activation, ROS generation and subsequent caspase-dependent apoptotic events. However, in the HSP25 overexpressed cells, the above-described effects were blocked. In fact, radiation-induced membrane translocation of PKCdelta and tyrosine phosphorylation were inhibited by HSP25. Based on the above data, we suggest that HSP25 downregulates PKCdelta, which is a key molecule for radiation-induced ROS generation and mitochondrial-mediated caspase-dependent apoptotic events.


Subject(s)
Apoptosis/radiation effects , Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Protein Kinase C/physiology , Reactive Oxygen Species/metabolism , Enzyme Activation , HSP27 Heat-Shock Proteins , Humans , Jurkat Cells , Mitochondria/physiology , Molecular Chaperones , Phosphorylation , Protein Kinase C-delta , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Cancer Res ; 63(20): 6825-37, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14583480

ABSTRACT

Epidemiological and experimental carcinogenesis studies provide evidence that components of garlic (Allium sativum) have anticancer activity. We recently reported that the garlic derivative S-allylmercaptocysteine (SAMC) inhibits growth, arrests cells in G(2)-M, and induces apoptosis in human colon cancer cells (Shirin et al., Cancer Res., 61: 725-731, 2001). Because a fraction of the SAMC-treated cells are specifically arrested in mitosis, we examined the mechanism of this effect in the present study. Immunofluorescent microscopy revealed that the treatment of SW480 cells or NIH3T3 fibroblasts with 150 micro M SAMC (the IC(50) concentration) caused rapid microtubule (MT) depolymerization, MT cytoskeleton disruption, centrosome fragmentation and Golgi dispersion in interphase cells. It also induced the formation of monopolar and multipolar spindles in mitotic cells. In vitro turbidity assays indicated that SAMC acted directly on tubulin to cause MT depolymerization, apparently because it interacts with -SH groups on tubulin. To investigate the signaling pathways involved in SAMC-induced apoptosis, we assayed c-Jun NH(2)-terminal kinase (JNK) activity and found that treatment with SAMC caused a rapid and sustained induction of JNK activity. The selective JNK inhibitor SP600125 inhibited the early phase (24 h) but not the late phase (48 h and later) of apoptosis induced by SAMC. Expression of a dominant-negative mutant of JNK1 in SW480 cells inhibited apoptosis induced by SAMC at 24 h but had no protective effect at 48 h. JNK1(-/-) mouse embryonic fibroblasts were resistant to SAMC-induced apoptosis at 24 h but not at 48 h. On the other hand, the inhibition or abrogation of JNK1 activity did not inhibit the G(2)-M arrest induced by SAMC. SAMC also activated caspase-3. The general caspase inhibitor z-VAD-fmk inhibited both early and late phases of apoptosis induced by SAMC. We conclude that the garlic-derived compound SAMC exerts antiproliferative effects by binding directly to tubulin and disrupting the MT assembly, thus arresting cells in mitosis and triggering JNK1 and caspase-3 signaling pathways that lead to apoptosis.


Subject(s)
Apoptosis/drug effects , Cysteine/analogs & derivatives , Cysteine/pharmacology , Microtubules/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/physiology , Binding Sites , Caspase 3 , Caspases/metabolism , Cell Cycle/physiology , Cell Line, Tumor , Centrosome/drug effects , Centrosome/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/physiology , Humans , Mice , Microtubules/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitosis/drug effects , Mitosis/physiology , NIH 3T3 Cells , Tubulin/metabolism , p38 Mitogen-Activated Protein Kinases
17.
Oncogene ; 23(56): 9062-9, 2004 Dec 02.
Article in English | MEDLINE | ID: mdl-15489897

ABSTRACT

We previously demonstrated that protein kinase C-eta (PKC-eta) mediates a phorbol 12-myristate-13-acetate (PMA)-induced proliferative response in human glioblastoma (GBM) cells. In this report, we show that PMA-stimulated activation of PKC-eta in U-251 GBM cells resulted in activation of both Akt and the mammalian target of rapamycin (mTOR) signaling pathways and an increase in cell proliferation. Expression of a kinase dead PKC-eta (PKC-etaKR) construct reduced the basal and PMA-evoked proliferation of PKC-eta-expressing U-251 GBM cells, as well as abrogated the PMA-induced activation of Akt, mTOR, and the mTOR targets 4E-BP1 and STAT-3. Treatment of cells with the PI-3 kinase inhibitor LY294002 (10 muM) or the mTOR inhibitor rapamycin (10 nM) also reduced PMA-induced proliferation and cell-cycle progression. Expression of a constitutively active PKC-eta (PKC-etaDeltaNPS) construct in a GBM cell line with no endogenous PKC-eta (U-1242) also provided evidence that PKC-eta targets the Akt and mTOR signaling pathways. Moreover, activation of 4E-BP1 and STAT-3 in both PMA-treated U-251 and PKC-etaDeltaNPS-expressing U-1242 GBM cells was inhibited by rapamycin. However, activation of Akt, but not mTOR was inhibited by the PI-3 kinase inhibitor LY294002. This study identifies Akt and mTOR as downstream targets of PKC-eta that are involved in GBM cell proliferation.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Kinase C/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Cell Cycle/drug effects , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Glioblastoma/enzymology , Glioblastoma/metabolism , Humans , Morpholines/pharmacology , Proto-Oncogene Proteins c-akt , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tetradecanoylphorbol Acetate/pharmacology
18.
Biochim Biophys Acta ; 1592(2): 205-11, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12379484

ABSTRACT

Protein kinase Cdelta (PKCdelta) is one of the functionally distinct isoforms in PKC family. p300 is a histone acetyltransferase/transcription coactivator. They share certain properties, such as ubiquitous expression, growth and tumor suppression, and ability to enhance differentiation and apoptosis. In this study, we found that PKCdelta but not classical PKC, specifically phosphorylates p300 at serine 89 in vitro and in vivo. This phosphorylation causes inhibition of p300 intrinsic HAT activity. Subsequently, the targeted acetylation of nucleosomal histones is markedly reduced, which causes repression of p300 transcription coactivator function. These findings identify a new signal transduction pathway by which PKCdelta may inhibit cell growth and promote cellular differentiation.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Isoenzymes/metabolism , Protein Kinase C/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Acetyltransferases/chemistry , Acetyltransferases/genetics , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , HeLa Cells , Histone Acetyltransferases , Humans , Phosphorylation , Protein Kinase C-delta , Serine/chemistry , Signal Transduction , Transcription Factors , Transfection , p300-CBP Transcription Factors
19.
Biochim Biophys Acta ; 1593(2-3): 219-29, 2003 Feb 17.
Article in English | MEDLINE | ID: mdl-12581866

ABSTRACT

Protein kinase C (PKC) isoforms play distinct roles in cellular functions. We have previously shown that ionizing radiation activates PKC isoforms (alpha, delta, epsilon, and zeta), however, isoform-specific sensitivities to radiation and its exact mechanisms in radiation mediated signal transduction are not fully understood. In this study, we showed that overexpression of PKC isoforms (alpha, delta, epsilon, and zeta) increased radiation-induced cell death in NIH3T3 cells and PKC epsilon overexpression was predominantly responsible. In addition, PKC epsilon overexpression increased ERK1/2 activation without altering other MAP-kinases such as p38 MAPK or JNK. Co-transfection of dominant negative PKC epsilon (PKC epsilon -KR) blocked both PKC epsilon -mediated ERK1/2 activation and radiation-induced cell death, while catalytically active PKC epsilon construction augmented these phenomena. When the PKC epsilon overexpressed cells were pretreated with PD98059, MEK inhibitor, radiation-induced cell death was inhibited. Co-transfection of the cells with a mutant of ERK1 or -2 (ERK1-KR or ERK2-KR) also blocked these phenomena, and co-transfection with dominant negative Ras or Raf cDNA revealed that PKC epsilon -mediated ERK1/2 activation was Ras-Raf-dependent. In conclusion, PKC epsilon -mediated ERK1/2 activation was responsible for the radiation-induced cell death.


Subject(s)
3T3 Cells/radiation effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , 3T3 Cells/metabolism , Animals , Cell Death/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Protein Kinase C/biosynthesis , Protein Kinase C/genetics , Protein Kinase C-epsilon , Proto-Oncogene Proteins c-raf/metabolism , Transfection , ras Proteins/metabolism
20.
Mol Cancer Ther ; 1(10): 803-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12492113

ABSTRACT

Recent studies provide evidence that exisulind and two potent derivatives, CP461 and CP248, induce apoptosis in colon cancer cells by inhibiting cyclic GMP (cGMP)-specific phosphodiesterases (phosphodiesterases 2 and 5). This causes an increase in intracellular levels of cGMP, thus activating the cGMP-dependent protein kinase G (PKG), which then activates pathways that lead to apoptosis. To further examine this mechanism and to provide a potential in vivo biomarker for activation of this pathway, we examined phosphorylation of the vasodilator-stimulated phosphoprotein (VASP), a ubiquitously expressed endogenous substrate for PKG. We found that VASP was phosphorylated after treating SW480 colon cancer cells with exisulind, CP461, or CP248. CP248-induced VASP phosphorylation was inhibited by a specific PKG inhibitor but not by a protein kinase A inhibitor. The drug 3-(5'-hydroxymethyl-2'-furyl)-benzylindazole and nitric oxide donors that activate cellular guanylyl cyclase and thus increase cellular levels of cGMP also caused VASP phosphorylation. With all of these agents, the phosphorylation of VASP was associated with increased intracellular levels of cGMP and the induction of apoptosis. We also demonstrated direct in vivo phosphorylation of VASP with constitutively activated mutants of PKG. These results suggest that VASP phosphorylation can provide a useful endogenous cellular biomarker for anticancer agents that cause cGMP-mediated apoptosis.


Subject(s)
Cell Adhesion Molecules/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Phosphoproteins/metabolism , Sulindac/analogs & derivatives , Sulindac/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers , Blotting, Western , COS Cells , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP/metabolism , Enzyme Activation , Enzyme Inhibitors , Humans , Microfilament Proteins , Mutation , Phosphodiesterase Inhibitors/pharmacology , Phosphorylation , Sulindac/pharmacology , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL