Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 6(2): e16803, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21347298

ABSTRACT

Volume-regulated anion channels (VRAC) are widely expressed chloride channels that are critical for the cell volume regulation. In the mammalian central nervous system, the physiological expression of neuronal VRAC and its role in cerebral ischemia are issues largely unknown. We show that hypoosmotic medium induce an outwardly rectifying chloride conductance in CA1 pyramidal neurons in rat hippocampal slices. The induced chloride conductance was sensitive to some of the VRAC inhibitors, namely, IAA-94 (300 µM) and NPPB (100 µM), but not to tamoxifen (10 µM). Using oxygen-and-glucose deprivation (OGD) to simulate ischemic conditions in slices, VRAC activation appeared after OGD induced anoxic depolarization (AD) that showed a progressive increase in current amplitude over the period of post-OGD reperfusion. The OGD induced VRAC currents were significantly inhibited by inhibitors for glutamate AMPA (30 µM NBQX) and NMDA (40 µM AP-5) receptors in the OGD solution, supporting the view that induction of AD requires an excessive Na(+)-loading via these receptors that in turn to activate neuronal VRAC. In the presence of NPPB and DCPIB in the post-OGD reperfusion solution, the OGD induced CA1 pyramidal neuron death, as measured by TO-PRO-3-I staining, was significantly reduced, although DCPIB did not appear to be an effective neuronal VRAC blocker. Altogether, we show that rat hippocampal pyramidal neurons express functional VRAC, and ischemic conditions can initial neuronal VRAC activation that may contribute to ischemic neuronal damage.


Subject(s)
CA1 Region, Hippocampal/pathology , Chloride Channels/metabolism , Glucose/deficiency , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Animals , Brain Ischemia/pathology , Cell Death , Male , Osmosis , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism , Reperfusion
2.
Endocr Res ; 34(1-2): 31-42, 2009.
Article in English | MEDLINE | ID: mdl-19557589

ABSTRACT

INTRODUCTION: In CV-1 cells, shuttling from cytoplasm to nucleus of the nuclear thyroid hormone receptor-beta1 (TRbeta1, TR) is shown in this report to be regulated by extracellular thyroid hormone at a hormone receptor on cell surface integrin alphav3. METHODS: The receptor was introduced into cells as a GFP-TR1 chimera and intracellular movement of the receptor was monitored by confocal microscopy of cells treated with L-thyroxine (T(4)). RESULTS AND DISCUSSION: TR-GFP translocation in the presence of T(4) requires activation of extracellular-regulated protein kinases 1/2 (ERK1/2). Inhibition of T(4)-binding to alphavbeta3 with anti-alphavbeta3 or Arg-Gly-Asp (RGD) peptide blocks T(4)-stimulated GFP-TR nuclear translocation, as do the hormone-binding inhibitor tetraiodothyroacetic acid (tetrac) and the ERK1/2 inhibitor, PD98059. TR1 is an ERK1/2 substrate. CONCLUSIONS: Via a nongenomic mechanism initiated at plasma membrane integrin v3, T(4)-activated ERK1/2 and TR1 move transiently in an immunoprecipitable complex to the nuclei of T(4)-treated cells.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Integrin alphaVbeta3/physiology , Receptors, Thyroid Hormone/physiology , Animals , Chlorocebus aethiops , Flavonoids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oligopeptides/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Thyroxine/analogs & derivatives , Thyroxine/pharmacology , Vero Cells
3.
J Cell Biochem ; 104(6): 2131-42, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18446786

ABSTRACT

Cyclooxygenase-2 (COX-2) content is increased in many types of tumor cells. We have investigated the mechanism by which resveratrol, a stilbene that is pro-apoptotic in many tumor cell lines, causes apoptosis in human head and neck squamous cell carcinoma UMSCC-22B cells by a mechanism involving cellular COX-2. UMSCC-22B cells treated with resveratrol for 24 h, with or without selected inhibitors, were examined: (1) for the presence of nuclear activated ERK1/2, p53 and COX-2, (2) for evidence of apoptosis, and (3) by chromatin immunoprecipitation to demonstrate p53 binding to the p21 promoter. Stilbene-induced apoptosis was concentration-dependent, and associated with ERK1/2 activation, serine-15 p53 phosphorylation and nuclear accumulation of these proteins. These effects were blocked by inhibition of either ERK1/2 or p53 activation. Resveratrol also caused p53 binding to the p21 promoter and increased abundance of COX-2 protein in UMSCC-22B cell nuclei. Resveratrol-induced nuclear COX-2 accumulation was dependent upon ERK1/2 activation, but not p53 activation. Activation of p53 and p53-dependent apoptosis were blocked by the COX-2 inhibitor, NS398, and by transfection of cells with COX-2-siRNA. In UMSCC-22B cells, resveratrol-induced apoptosis and induction of nuclear COX-2 accumulation share dependence on the ERK1/2 signal transduction pathway. Resveratrol-inducible nuclear accumulation of COX-2 is essential for p53 activation and p53-dependent apoptosis in these cancer cells.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Head and Neck Neoplasms/enzymology , Neoplasms, Squamous Cell/enzymology , Stilbenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Head and Neck Neoplasms/pathology , Humans , MAP Kinase Signaling System/drug effects , Models, Biological , Neoplasms, Squamous Cell/pathology , Nitrobenzenes/pharmacology , Phosphoserine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Resveratrol , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology , Sulfonamides/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cancer Ther ; 5(8): 2034-42, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928824

ABSTRACT

Cyclooxygenase-2 (COX-2) is antiapoptotic and is implicated in tumorigenesis. Recent reports, however, have also ascribed a proapoptotic action to inducible COX-2. We show here for the first time that a stilbene, resveratrol, induces nuclear accumulation of COX-2 protein in human breast cancer MCF-7 and MDA-MB-231 cell cultures. The induction of COX-2 accumulation by resveratrol is mitogen-activated protein kinase (MAPK; extracellular signal-regulated kinase 1/2)- and activator protein 1- dependent. Nuclear COX-2 in resveratrol-treated cells colocalizes with Ser(15)-phosphorylated p53 and with p300, a coactivator for p53-dependent gene expression. The interaction of COX-2, p53, and p300, as well as resveratrol-induced apoptosis, was inhibited by a MAPK activation inhibitor, PD98059. A specific inhibitor of COX-2, NS398, and small interfering RNA knockdown of COX-2 were associated with reduced p53 phosphorylation and consequent decrease in p53-dependent apoptosis in resveratrol-treated cells. We conclude that nuclear accumulation of COX-2 can be induced by resveratrol and that the COX has a novel intranuclear colocalization with Ser(15)-phosphorylated p53 and p300, which facilitates apoptosis in resveratrol-treated breast cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cyclooxygenase 2/drug effects , Stilbenes/pharmacology , Apoptosis/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Nucleus/metabolism , Cyclooxygenase 2/metabolism , E1A-Associated p300 Protein/drug effects , E1A-Associated p300 Protein/metabolism , Female , Humans , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Resveratrol , Serine/metabolism , Signal Transduction , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
5.
FASEB J ; 20(10): 1742-4, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16790523

ABSTRACT

Resveratrol is a naturally occurring polyphenol, which causes apoptosis in cultured cancer cells. We describe a cell surface resveratrol receptor on the extracellular domain of hetero-dimeric alphaVbeta3 integrin in MCF-7 human breast cancer cells. This receptor is linked to induction by resveratrol of extracellular-regulated kinases 1 and 2 (ERK1/2)- and serine-15-p53-dependent phosphorylation leading to apoptosis. The integrin receptor is near the Arg-Gly-Asp (RGD) recognition site on the integrin; an integrin-binding RGD peptide inhibits induction by resveratrol of ERK1/2- and p53-dependent apoptosis. Antibody (Ab) to integrin alphaVbeta3, but not to alphaVbeta5, inhibits activation by resveratrol of ERK1/2 and p53 and consequent apoptosis in estrogen receptor-alpha (ERalpha) positive MCF-7, and ERalpha-negative MDA-MB231 cells. Resveratrol is displaced from the purified integrin by an RGD, but not RGE, peptide, and by alphaVbeta3 integrin-specific Ab. Resveratrol action is blocked by siRNAbeta3, but not by siRNAalphaV. [14C]-Resveratrol binds to commercially purified integrin alphaVbeta3 and to alphaVbeta3 prepared from MCF-7 cells; binding of [14C]-resveratrol to the beta3, but not to the alphaV monomer, is displaced by unlabeled resveratrol. In conclusion, binding of resveratrol to integrin alphaVbeta3, principally to the beta3 monomer, is essential for transduction of the stilbene signal into p53-dependent apoptosis of breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Stilbenes/pharmacokinetics , Apoptosis/drug effects , Binding Sites , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Humans , Integrin beta3/genetics , Integrin beta3/metabolism , Resveratrol , Signal Transduction/drug effects , Stilbenes/pharmacology , Tumor Suppressor Protein p53
6.
Steroids ; 70(5-7): 444-9, 2005.
Article in English | MEDLINE | ID: mdl-15862828

ABSTRACT

Because the androgen and estrogen nuclear hormone receptors are subject to acetylation, we speculated that the nuclear thyroid hormone receptor-beta1 (TRbeta1), another superfamily member, was also subject to this posttranslational modification. Treatment of 293T cells that contain TRbeta1(wt) with l-thyroxine (T4)(10(-7)M, total concentration) resulted in the accumulation of acetylated TR in nuclear fractions at 30-45 min and a decrease in signal by 60 min. A similar time course characterized recruitment by TR of p300, a coactivator protein with intrinsic transacetylase activity. Recruitment by the receptor of SRC-1, a TR coactivator that also acetylates nucleoproteins, was also demonstrated. Inhibition of the MAPK (ERK1/2) signal transduction cascade by PD 98059 blocked the acetylation of TR caused by T4. Tetraiodothyroacetic acid (tetrac) decreased T4-induced acetylation of TR. At 10(-7)M, 3,5,3'-triiodo-l-thyronine (T3) was comparably effective to T4 in causing acetylation of TR. We studied acetylation in TR that contained mutations in the DNA-binding domain (DBD) (residues 128-142) that are known to be relevant to recruitment of coactivators and to include the MAPK docking site. In response to T4 treatment, the K128A TR mutant transfected into CV-1 cells recruited p300, but not SRC-1, and was subject to acetylation. R132A complexed with SRC-1, but not p300; it was acetylated equally well in both the absence and presence of T4. S142E was acetylated in the absence and presence of T4 and bound SRC-1 under both conditions; this mutant was also capable of binding p300 in the presence of T4. There was no serine phosphorylation of TR in any of these mutants. We conclude that (1) TRbeta1, like AR and ER, is subject to acetylation; (2) the process of acetylation of TR requires thyroid hormone-directed MAPK activity, but not serine phosphorylation of TR by MAPK, suggesting that the contribution of MAPK is upstream in the activation of the acetylase; (3) the amino acid residue 128-142 region of the DBD of TR is important to thyroid hormone-associated recruitment of p300 and SRC-1; (4) acetylation of TR DBD mutants that is directed by T4 appears to be associated with recruitment of p300.


Subject(s)
MAP Kinase Signaling System , Receptors, Cytoplasmic and Nuclear/metabolism , Thyroid Hormones/metabolism , Acetylation , Animals , Cell Line , Cell Nucleus/metabolism , Enzyme Activation , Flavonoids/pharmacology , Humans , Mutation , Phosphorylation , Plasmids/metabolism , Protein Structure, Tertiary , Receptors, Thyroid Hormone/metabolism , Serine/chemistry , Signal Transduction , Subcellular Fractions/metabolism , Thyroid Hormone Receptors beta , Thyroxine/metabolism , Time Factors , Transfection , Tumor Suppressor Protein p53/metabolism
7.
Mol Cancer Ther ; 3(11): 1355-64, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542774

ABSTRACT

Resveratrol, a naturally occurring stilbene with antitumor properties, caused mitogen-activated protein kinase [MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2)] activation, nuclear translocation of Ser15-phosphorylated p53, and p53-dependent apoptosis in hormone-insensitive DU145 prostate cancer cells. Exposure of these cells to epidermal growth factor (EGF) for up to 4 hours resulted in brief activation of MAPK followed by inhibition of resveratrol-induced signal transduction, p53 phosphorylation, and apoptosis. Resveratrol stimulated c-fos and c-jun expression in DU145 cells, an effect also suppressed by EGF. An inhibitor of protein kinase C (PKC)-alpha, -beta, and -gamma (CGP41251) enhanced Ser15 phosphorylation of p53 by resveratrol in the absence of EGF and blocked EGF inhibition of the resveratrol effect. EGF caused PKC-alpha/beta phosphorylation in DU145 cells, an effect reversed by CGP41251. Activation of PKC by phorbol ester (phorbol 12-myristate 13-acetate) enhanced EGF action on ERK1/2 phosphorylation without significantly altering p53 phosphorylation by resveratrol. DU145 cells transfected with a dominant-negative PKC-alpha construct showed resveratrol-induced ERK1/2 phosphorylation and Ser15 phosphorylation of p53 but were unresponsive to EGF. Thus, resveratrol and EGF activate MAPK by discrete mechanisms in DU145 cells. The stilbene promoted p53-dependent apoptosis, whereas EGF opposed induction of apoptosis by resveratrol via a PKC-alpha-mediated mechanism. Resveratrol also induced p53 phosphorylation in LNCaP prostate cancer cells, an effect also inhibited by EGF. Inhibition of PKC activation in LNCaP cells, however, resulted in a reduction, rather than increase, in p53 activation and apoptosis, suggesting that resveratrol-induced apoptosis in these two cell lines occurs through different PKC-mediated and MAPK-dependent pathways.


Subject(s)
Apoptosis/drug effects , Epidermal Growth Factor/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , Stilbenes/antagonists & inhibitors , Stilbenes/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Male , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Resveratrol , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
8.
Circ Res ; 94(11): 1500-6, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15117822

ABSTRACT

The effects of thyroid hormone analogues on modulation of angiogenesis have been studied in the chick chorioallantoic membrane model. Generation of new blood vessels from existing vessels was increased 3-fold by either l-thyroxine (T4; 10(-7) mol/L) or 3,5,3'-triiodo-l-thyronine (10(-9) mol/L). T4-agarose reproduced the effects of T4, and tetraiodothyroacetic acid (tetrac) inhibited the effects of both T4 and T4-agarose. Tetrac itself was inactive and is known to block actions of T4 on signal transduction that are initiated at the plasma membrane. T4 and basic fibroblast growth factor (FGF2) were comparably effective as inducers of angiogenesis. Low concentrations of FGF2 combined with submaximal concentrations of T4 produced an additive angiogenic response. Anti-FGF2 inhibited the angiogenic effect of T4. The proangiogenic effects of T4 and FGF2 were blocked by PD 98059, a mitogen-activated protein kinase (MAPK) pathway inhibitor. Endothelial cells (ECV304) treated with T4 or FGF2 for 15 minutes demonstrated activation of MAPK, an effect inhibited by PD 98059 and the protein kinase C inhibitor CGP41251. Reverse transcription-polymerase chain reaction of RNA extracted from endothelial cells treated with T4 revealed increased abundance of FGF2 transcript at 6 to 48 hours, and after 72 hours, the medium of treated cells showed increased FGF2 content, an effect inhibited by PD 98059. Thus, thyroid hormone is shown to be a proangiogenic factor. This action, initiated at the plasma membrane, is MAPK dependent and mediated by FGF2.


Subject(s)
Chorioallantoic Membrane/blood supply , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/physiology , MAP Kinase Signaling System/physiology , Neovascularization, Physiologic/drug effects , Thyroxine/analogs & derivatives , Thyroxine/pharmacology , Animals , Cell Membrane/physiology , Chick Embryo , Endothelial Cells/ultrastructure , Endothelium, Vascular/embryology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Humans , MAP Kinase Signaling System/drug effects , Neovascularization, Physiologic/physiology
9.
Endocrinology ; 145(4): 1708-17, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14691008

ABSTRACT

Epidermal growth factor (EGF) and TGFalpha share the same plasma membrane receptor. In the present studies in HeLa cells, both EGF and TGFalpha caused MAPK (ERK1/2) activation and expression of the immediate-early gene c-fos. Thyroid hormone (T(4)) nongenomically enhanced EGF- and TGFalpha-induced MAPK activation. This T(4) action was duplicated by T(4)-agarose and blocked by tetraiodothyroacetic acid, which inhibits binding of T(4) to plasma membranes. TGFalpha-induced MAPK activation was potentiated by 8-bromo-cyclic adenosine monophosphate (8-Br-cAMP) but not 8-chloro-cyclic adenosine monophosphate. TGFalpha, T(4), and 8-Br-cAMP each caused protein kinase A (PKA) II serine phosphorylation, whereas phosphorylation of PKA-II was not seen in cells treated with EGF or 8-chloro-cyclic adenosine monophosphate. In a PKA activity assay, the enzyme was stimulated by T(4), EGF, and TGFalpha; T(4) enhanced the effect of TGFalpha but not that of EGF. T(4), although it potentiated c-fos gene expression in EGF-treated cells, suppressed this effect in cells treated with TGFalpha. Cells exposed to 8-Br-cAMP also inhibited TGFalpha-stimulated c-fos expression. Studies of cell proliferation indicated that T(4) potentiated EGF action but inhibited that effect in TGFalpha-treated cells. The disparate effects of T(4) on actions of EGF and TGFalpha, which share the same cell surface receptor, are mediated by hormone phosphorylation and activation of PKA-II.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Epidermal Growth Factor/physiology , Thyroxine/physiology , Transforming Growth Factor alpha/physiology , 3T3 Cells , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Synergism , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Gene Expression Regulation/drug effects , Genes, Immediate-Early , HeLa Cells , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Thyroxine/pharmacology , Transforming Growth Factor alpha/pharmacology
10.
Am J Physiol Cell Physiol ; 284(6): C1429-37, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12519748

ABSTRACT

Human orbital fibroblasts are more susceptible to some actions of proinflammatory cytokines than are fibroblasts from other anatomic regions. These cells produce high levels of PGE(2) when activated by cytokines. Here we report that they express high levels of prostaglandin-endoperoxide H synthase (PGHS)-2, the inflammatory cyclooxygenase, when treated with IL-1beta. This induction results from enhanced PGHS-2 mRNA stability and small increases in gene promoter activity. The enhanced transcript stability is a result of actions of the cytokine on the 3'-untranslated region. Orbital fibroblasts, unlike those from skin, fail to express high levels of IL-1 receptor antagonist (IL-1ra) when treated with IL-1beta, leading to loss of modulation of IL-1 action. This can be overcome by transiently transfecting cells with IL-1ra. Thus a decreased level of IL-1ra expression in orbital fibroblasts may underlie the exaggerated responses to IL-1 observed in those cells and, therefore, the susceptibility of the orbit to inflammation.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Interleukin-1/metabolism , Isoenzymes/metabolism , Orbit , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Interleukin-1/metabolism , Sialoglycoproteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Cyclooxygenase 2 , Epidermal Cells , Epidermis/metabolism , Fibroblasts/cytology , Humans , Interleukin 1 Receptor Antagonist Protein , Isoenzymes/genetics , Membrane Proteins , NF-kappa B/metabolism , Promoter Regions, Genetic , Prostaglandin-Endoperoxide Synthases/genetics , Protein Binding , Protein Isoforms/metabolism , RNA Stability , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...