Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Cell Signal ; 36: 176-188, 2017 08.
Article in English | MEDLINE | ID: mdl-28495590

ABSTRACT

Activation of ß2-adrenergic receptor (ß2AR) and deorphanized GPR55 has been shown to modulate cancer growth in diverse tumor types in vitro and in xenograft models in vivo. (R,R')-4'-methoxy-1-naphthylfenoterol [(R,R')-MNF] is a bivalent compound that agonizes ß2AR but inhibits GPR55-mediated pro-oncogenic responses. Here, we investigated the molecular mechanisms underlying the anti-tumorigenic effects of concurrent ß2AR activation and GPR55 blockade in C6 glioma cells using (R,R')-MNF as a marker ligand. Our data show that (R,R')-MNF elicited G1-phase cell cycle arrest and apoptosis, reduced serum-inducible cell motility, promoted the phosphorylation of PKA target proteins, and inhibited constitutive activation of ERK and AKT in the low nanomolar range, whereas high nanomolar levels of (R,R')-MNF were required to block GPR55-mediated cell motility. siRNA knockdown and pharmacological inhibition of ß2AR activity were accompanied by significant upregulation of AKT and ERK phosphorylation, and selective alteration in (R,R')-MNF responsiveness. The effects of agonist stimulation of GPR55 on various readouts, including cell motility assays, were suppressed by (R,R')-MNF. Lastly, a significant increase in phosphorylation-mediated inactivation of ß-catenin occurred with (R,R')-MNF, and we provided new evidence of (R,R')-MNF-mediated inhibition of oncogenic ß-catenin signaling in a C6 xenograft tumor model. Thus, simultaneous activation of ß2AR and blockade of GPR55 may represent a novel therapeutic approach to combat the progression of glioblastoma cancer.


Subject(s)
Brain Neoplasms/metabolism , Carcinogenesis/metabolism , Glioma/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Cannabinoid/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Brain Neoplasms/pathology , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colforsin/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fenoterol/analogs & derivatives , Fenoterol/pharmacology , Glioma/pathology , Humans , Isoproterenol/pharmacology , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , Rats , Serum , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
2.
Cell Signal ; 27(5): 997-1007, 2015 May.
Article in English | MEDLINE | ID: mdl-25703025

ABSTRACT

(R,R')-4'-methoxy-1-naphthylfenoterol [(R,R')-MNF] is a highly-selective ß2 adrenergic receptor (ß2-AR) agonist. Incubation of a panel of human-derived melanoma cell lines with (R,R')-MNF resulted in a dose- and time-dependent inhibition of motility as assessed by in vitro wound healing and xCELLigence migration and invasion assays. Activity of (R,R')-MNF positively correlated with the ß2-AR expression levels across tested cell lines. The anti-motility activity of (R,R')-MNF was inhibited by the ß2-AR antagonist ICI-118,551 and the protein kinase A inhibitor H-89. The adenylyl cyclase activator forskolin and the phosphodiesterase 4 inhibitor Ro 20-1724 mimicked the ability of (R,R')-MNF to inhibit migration of melanoma cell lines in culture, highlighting the importance of cAMP for this phenomenon. (R,R')-MNF caused significant inhibition of cell growth in ß2-AR-expressing cells as monitored by radiolabeled thymidine incorporation and xCELLigence system. The MEK/ERK cascade functions in cellular proliferation, and constitutive phosphorylation of MEK and ERK at their active sites was significantly reduced upon ß2-AR activation with (R,R')-MNF. Protein synthesis was inhibited concomitantly both with increased eEF2 phosphorylation and lower expression of tumor cell regulators, EGF receptors, cyclin A and MMP-9. Taken together, these results identified ß2-AR as a novel potential target for melanoma management, and (R,R')-MNF as an efficient trigger of anti-tumorigenic cAMP/PKA-dependent signaling in ß2-AR-expressing lesions.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Fenoterol/analogs & derivatives , Melanoma/drug therapy , Receptors, Adrenergic, beta-2/metabolism , Cell Line, Tumor , Cyclic AMP/metabolism , Fenoterol/pharmacology , Humans , Melanoma/metabolism , Melanoma/pathology , Signal Transduction/drug effects
3.
Aging Cell ; 14(3): 334-44, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25711920

ABSTRACT

Astrocytes are key cells in brain aging, helping neurons to undertake healthy aging or otherwise letting them enter into a spiral of neurodegeneration. We aimed to characterize astrocytes cultured from senescence-accelerated prone 8 (SAMP8) mice, a mouse model of brain pathological aging, along with the effects of caloric restriction, the most effective rejuvenating treatment known so far. Analysis of the transcriptomic profiles of SAMP8 astrocytes cultured in control conditions and treated with caloric restriction serum was performed using mRNA microarrays. A decrease in mitochondrial and ribosome mRNA, which was restored by caloric restriction, confirmed the age-related profile of SAMP8 astrocytes and the benefits of caloric restriction. An amelioration of antioxidant and neurodegeneration-related pathways confirmed the brain benefits of caloric restriction. Studies of oxidative stress and mitochondrial function demonstrated a reduction of oxidative damage and partial improvement of mitochondria after caloric restriction. In summary, caloric restriction showed a significant tendency to normalize pathologically aged astrocytes through the activation of pathways that are protective against the age-related deterioration of brain physiology.


Subject(s)
Aging/metabolism , Astrocytes/metabolism , Caloric Restriction , Animals , Antioxidants/metabolism , Caloric Restriction/methods , Cells, Cultured , Mice , Mitochondria/metabolism , Neurons/metabolism , Oxidative Stress/physiology
4.
Anesthesiology ; 121(1): 149-59, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24936922

ABSTRACT

BACKGROUND: Subanesthetic doses of (R,S)-ketamine are used in the treatment of neuropathic pain and depression. In the rat, the antidepressant effects of (R,S)-ketamine are associated with increased activity and function of mammalian target of rapamycin (mTOR); however, (R,S)-ketamine is extensively metabolized and the contribution of its metabolites to increased mTOR signaling is unknown. METHODS: Rats (n = 3 per time point) were given (R,S)-ketamine, (R,S)-norketamine, and (2S,6S)-hydroxynorketamine and their effect on the mTOR pathway determined after 20, 30, and 60 min. PC-12 pheochromocytoma cells (n = 3 per experiment) were treated with escalating concentrations of each compound and the impact on the mTOR pathway was determined. RESULTS: The phosphorylation of mTOR and its downstream targets was significantly increased in rat prefrontal cortex tissue by more than ~2.5-, ~25-, and ~2-fold, respectively, in response to a 60-min postadministration of (R,S)-ketamine, (R,S)-norketamine, and (2S,6S)-hydroxynorketamine (P < 0.05, ANOVA analysis). In PC-12 pheochromocytoma cells, the test compounds activated the mTOR pathway in a concentration-dependent manner, which resulted in a significantly higher expression of serine racemase with ~2-fold increases at 0.05 nM (2S,6S)-hydroxynorketamine, 10 nM (R,S)-norketamine, and 1,000 nM (R,S)-ketamine. The potency of the effect reflected antagonistic activity of the test compounds at the α7-nicotinic acetylcholine receptor. CONCLUSIONS: The data demonstrate that (R,S)-norketamine and (2S,6S)-hydroxynorketamine have potent pharmacological activity both in vitro and in vivo and contribute to the molecular effects produced by subanesthetic doses of (R,S)-ketamine. The results suggest that the determination of the mechanisms underlying the antidepressant and analgesic effects of (R,S)-ketamine requires a full study of the parent compound and its metabolites.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Ketamine/analogs & derivatives , TOR Serine-Threonine Kinases/drug effects , Aconitine/analogs & derivatives , Aconitine/pharmacology , Animals , Blotting, Western , Brain/metabolism , Ketamine/analysis , Ketamine/pharmacokinetics , Ketamine/pharmacology , Male , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , PC12 Cells , Phosphorylation , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
5.
J Biol Chem ; 289(28): 19351-63, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24831005

ABSTRACT

Interaction of a given G protein-coupled receptor to multiple different G proteins is a widespread phenomenon. For instance, ß2-adrenoceptor (ß2-AR) couples dually to Gs and Gi proteins. Previous studies have shown that cAMP-dependent protein kinase (PKA)-mediated phosphorylation of ß2-AR causes a switch in receptor coupling from Gs to Gi. More recent studies have demonstrated that phosphorylation of ß2-AR by G protein-coupled receptor kinases, particularly GRK2, markedly enhances the Gi coupling. We have previously shown that although most ß2-AR agonists cause both Gs and Gi activation, (R,R')-fenoterol preferentially activates ß2-AR-Gs signaling. However, the structural basis for this functional selectivity remains elusive. Here, using docking simulation and site-directed mutagenesis, we defined Tyr-308 as the key amino acid residue on ß2-AR essential for Gs-biased signaling. Following stimulation with a ß2-AR-Gs-biased agonist (R,R')-4'-aminofenoterol, the Gi disruptor pertussis toxin produced no effects on the receptor-mediated ERK phosphorylation in HEK293 cells nor on the contractile response in cardiomyocytes expressing the wild-type ß2-AR. Interestingly, Y308F substitution on ß2-AR enabled (R,R')-4'-aminofenoterol to activate Gi and to produce these responses in a pertussis toxin-sensitive manner without altering ß2-AR phosphorylation by PKA or G protein-coupled receptor kinases. These results indicate that, in addition to the phosphorylation status, the intrinsic structural feature of ß2-AR plays a crucial role in the receptor coupling selectivity to G proteins. We conclude that specific interactions between the ligand and the Tyr-308 residue of ß2-AR stabilize receptor conformations favoring the receptor-Gs protein coupling and subsequently result in Gs-biased agonism.


Subject(s)
Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Adrenergic beta-2 Receptor Agonists/pharmacology , Amino Acid Substitution , Animals , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Mutation, Missense , Myocytes, Cardiac/cytology , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-2/genetics , Signal Transduction/drug effects , Tyrosine/genetics , Tyrosine/metabolism
6.
Biochem Pharmacol ; 87(4): 547-61, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24355564

ABSTRACT

(R,R')-4'-Methoxy-1-naphthylfenoterol (MNF) promotes growth inhibition and apoptosis of human HepG2 hepatocarcinoma cells via cannabinoid receptor (CBR) activation. The synthetic CB1R inverse agonist, AM251, has been shown to block the anti-mitogenic effect of MNF in these cells; however, AM251 is also an agonist of the recently deorphanized, lipid-sensing receptor, GPR55, whose upregulation contributes to carcinogenesis. Here, we investigated the role of MNF in GPR55 signaling in human HepG2 and PANC-1 cancer cell lines in culture by focusing first on internalization of the fluorescent ligand Tocrifluor 1117 (T1117). Initial results indicated that cell pretreatment with GPR55 agonists, including the atypical cannabinoid O-1602 and l-α-lysophosphatidylinositol, dose-dependently reduced the rate of cellular T1117 uptake, a process that was sensitive to MNF inhibition. GPR55 internalization and signaling mediated by O-1602 was blocked by MNF in GPR55-expressing HEK293 cells. Pretreatment of HepG2 and PANC-1 cells with MNF significantly abrogated the induction of ERK1/2 phosphorylation in response to AM251 and O-1602. Moreover, MNF exerted a coordinated negative regulation of AM251 and O-1602 inducible processes, including changes in cellular morphology and cell migration using scratch wound healing assay. This study shows for the first time that MNF impairs GPR55-mediated signaling and, therefore, may have therapeutic potential in the management of cancer.


Subject(s)
Cell Movement/drug effects , Drug Inverse Agonism , Endocytosis/physiology , Fenoterol/analogs & derivatives , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Cell Movement/physiology , Endocytosis/drug effects , Fenoterol/administration & dosage , HEK293 Cells , Hep G2 Cells , Humans , Ligands , Piperidines/administration & dosage , Piperidines/chemistry , Piperidines/metabolism , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Pyrazoles/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/physiology , Receptors, Cannabinoid , Receptors, G-Protein-Coupled/metabolism
7.
Cell Signal ; 25(12): 2634-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24012499

ABSTRACT

Western blot analysis demonstrated that PC-12 cells express monomeric and dimeric forms of serine racemase (m-SR, d-SR) and that 1321N1 cells express m-SR. Quantitative RT-PCR and functional studies demonstrated that PC-12 cells express homomeric and heteromeric forms of nicotinic acetylcholine receptors (nAChR) while 1321N1 cells primarily express the α7-nAChR subtype. The effect of nAChR agonists and antagonists on SR activity and expression was examined by following concentration-dependent changes in intracellular d-Ser levels and SR protein expression. Incubation with (S)-nicotine increased d-Ser levels, which were attenuated by the α7-nAChR antagonist methyllycaconitine (MLA). Treatment of PC-12 cells with mecamylamine (MEC) produced a bimodal reduction of d-Ser reflecting MEC inhibition of homomeric and heteromeric nAChRs, while a unimodal curve was observed with 1321N1 cells, reflecting predominant expression of α7-nAChR. The nAChR subtype selectivity was probed using α7-nAChR selective inhibitors MLA and (R,S)-dehydronorketamine and α3ß4-nAChR specific inhibitor AT-1001. The compounds reduced d-Ser in PC-12 cells, but only MLA and (R,S)-dehydronorketamine were effective in 1321N1 cells. Incubation of PC-12 and 1321N1 cells with (S)-nicotine, MEC and AT-1001 did not affect m-SR or d-SR expression, while MLA and (R,S)-dehydronorketamine increased m-SR expression but not SR mRNA levels. Treatment with cycloheximide indicated that increased m-SR was due to de novo protein synthesis associated with phospho-active forms of ERK1/2, MARCKS, Akt and rapamycin-sensitive mTOR. This effect was attenuated by treatment with the pharmacological inhibitors U0126, LY294002 and rapamycin, which selectively block the activation of ERK1/2, Akt and mTOR, respectively, and siRNAs directed against ERK1/2, Akt and mTOR. We propose that nAChR-associated changes in Ca(2+) flux affect SR activity, but not expression, and that MLA and (R,S)-dehydronorketamine bind to allosteric sites on the α7-nAChR and promote multiple signaling cascades that converge at mTOR to increase m-SR levels.


Subject(s)
Aconitine/analogs & derivatives , Mecamylamine/pharmacology , Nicotinic Antagonists/pharmacology , PC12 Cells/drug effects , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Receptors, Nicotinic/metabolism , Aconitine/pharmacology , Animals , Cell Line , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Nicotine/metabolism , PC12 Cells/enzymology , PC12 Cells/metabolism , Rats , Receptors, Nicotinic/genetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Neurosci Lett ; 535: 90-4, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23274708

ABSTRACT

The effects of gabapentin (GBP) and (S)-pregabalin (PGB) on the intracellular concentrations of d-serine and the expression of serine racemase (SR) in PC-12 cells were determined. Intracellular d-serine concentrations were determined using an enantioselective capillary electrophoresis assay with laser-induced fluorescence detection. Increasing concentrations of GBP, 0.1-20µM, produced a significant decrease in d-serine concentration relative to control, 22.9±6.7% at 20µM (*p<0.05), with an IC(50) value of 3.40±0.29µM. Increasing concentrations of PGB, 0.1-10µM, produced a significant decrease in d-serine concentration relative to control, 25.3±7.6% at 10µM (*p<0.05), with an IC(50) value of 3.38±0.21µM. The compounds had no effect on the expression of monomeric-SR or dimeric-SR as determined by Western blotting. The results suggest that incubation of PC-12 cells with GBP and PGB reduced the basal activity of SR, which is most likely a result of the decreased Ca(2+) flux produced via interaction of the drugs with the α(2)-δ subunit of voltage-gated calcium channels. d-Serine is a co-agonist of the N-methyl d-aspartate receptor (NMDAR) and reduced d-serine concentrations have been associated with reduced NMDAR activity. Thus, GBP and PGB may act as indirect antagonists of NMDAR, a mechanism that may contribute to the clinical effects of the drugs in neuropathic pain.


Subject(s)
Amines/pharmacology , Analgesics/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Serine/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Gabapentin , Intracellular Space/metabolism , PC12 Cells , Pregabalin , Racemases and Epimerases/metabolism , Rats , Serine/chemistry , Stereoisomerism , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/pharmacology
9.
Pharmacol Res Perspect ; 1(2): e00010, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25505565

ABSTRACT

(R,R')-4-methoxy-1-naphthylfenoterol (MNF) inhibits cancer cell proliferation in vitro through cell-type specific modulation of ß2-adrenergic receptor and/or cannabinoid receptor function. Here, we report an investigation into antitumor activity of MNF in rat C6 glioma cells. The potent antiproliferative action of MNF in these cells (IC50 of ∼1 nmol/L) was refractory to pharmacological inhibition of ß2-adrenergic receptor while a synthetic inverse agonist of cannabinoid receptor 1 significantly blocked MNF activity. The antitumor activity of MNF was then assessed in a C6 glioblastoma xenograft model in mice. Three days after subcutaneous implantation of C6 cells into the lower flank of nude mice, these animals were subjected to i.p. injections of saline or MNF (2 mg/kg) for 19 days and tumor volumes were measured over the course of the experiment. Gene expression analysis, quantitative RT-PCR and immunoblot assays were performed on the tumors after treatment. Significant reduction in mean tumor volumes was observed in mice receiving MNF when compared with the saline-treated group. We identified clusters in expression of genes involved in cellular proliferation, as well as molecular markers for glioblastoma that were significantly downregulated in tumors of MNF-treated mice as compared to saline-injected controls. The efficacy of MNF against C6 glioma cell proliferation in vivo and in vitro was accompanied by marked reduction in the expression of cell cycle regulator proteins. This study is the first demonstration of MNF-dependent chemoprevention of a glioblastoma xenograft model and may offer a potential mechanism for its anticancer action in vivo.

10.
J Pharmacol Exp Ther ; 343(1): 157-66, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22776956

ABSTRACT

Inhibition of cell proliferation by fenoterol and fenoterol derivatives in 1321N1 astrocytoma cells is consistent with ß(2)-adrenergic receptor (ß(2)-AR) stimulation. However, the events that result in fenoterol-mediated control of cell proliferation in other cell types are not clear. Here, we compare the effect of the ß(2)-AR agonists (R,R')-fenoterol (Fen) and (R,R')-4-methoxy-1-naphthylfenoterol (MNF) on signaling and cell proliferation in HepG2 hepatocarcinoma cells by using Western blotting and [(3)H]thymidine incorporation assays. Despite the expression of ß(2)-AR, no cAMP accumulation was observed when cells were stimulated with isoproterenol or Fen, although the treatment elicited both mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt activation. Unexpectedly, isoproterenol and Fen promoted HepG2 cell growth, but MNF reduced proliferation together with increased apoptosis. The mitogenic responses of Fen were attenuated by 3-(isopropylamino)-1-[(7-methyl-4-indanyl)oxy]butan-2-ol (ICI 118,551), a ß(2)-AR antagonist, whereas those of MNF were unaffected. Because of the coexpression of ß(2)-AR and cannabinoid receptors (CBRs) and their impact on HepG2 cell proliferation, these Gα(i)/Gα(o)-linked receptors may be implicated in MNF signaling. Cell treatment with (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-napthalenylmethanone (WIN 55,212-2), a synthetic agonist of CB(1)R and CB(2)R, led to growth inhibition, whereas inverse agonists of these receptors blocked MNF mitogenic responses without affecting Fen signaling. MNF responses were sensitive to pertussis toxin. The ß(2)-AR-deficient U87MG cells were refractory to Fen, but responsive to the antiproliferative actions of MNF and WIN 55,212-2. The data indicate that the presence of the naphthyl moiety in MNF results in functional coupling to the CBR pathway, providing one of the first examples of a dually acting ß(2)-AR-CBR ligand.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Apoptosis/physiology , Carcinoma, Hepatocellular/metabolism , Fenoterol/pharmacology , Liver Neoplasms/metabolism , Receptors, Cannabinoid/metabolism , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Fenoterol/chemistry , Hep G2 Cells , Humans , Liver Neoplasms/pathology
11.
Anal Biochem ; 421(2): 460-6, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22037294

ABSTRACT

An enantioselective capillary electrophoresis-laser-induced fluorescence (CE-LIF) method for the analysis of D-serine (D-Ser) in cellular matrices has been developed. The assay involves derivatization with FITC followed by CE-LIF using 0.5 mM hydroxyl propyl-ß-cyclodextrin in borate buffer [80 mM, pH 9.3]. The method was able to resolve D-Ser and L-Ser with an enantioselectivity (α) of 1.03 and a resolution (R(s)) of 1.37. Linearity was established from 0.25 to 100.00 µM. The assay was also able to enantioselectively resolve 6 additional amino acid racemates. The method was applied to the determination of intracellular D-Ser concentrations in PC-12, C6, 1312N1, and HepG2 cell lines. This method was used to determine the concentration-dependent increases in D-Ser and associated EC50 values produced by L-Ser and the concentration-dependent decreases in d-Ser and associated IC50 values produced by glycine, a competitive inhibitor of serine racemase (SR). Western blot analysis determined that the PC-12 and C6 cell lines contained monomeric and dimeric forms of SR while the 1321N1 and HepG2 cells contained only the monomeric form. Although the SR dimer has been identified as the active form of the enzyme, all four of the tested cell lines expressed enzymatically active SR.


Subject(s)
Electrophoresis, Capillary/methods , Racemases and Epimerases/analysis , Serine/analysis , Spectrometry, Fluorescence/methods , Animals , Blotting, Western , Hep G2 Cells , Humans , Lasers , Reference Standards
12.
J Biol Chem ; 286(22): 19270-9, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21467030

ABSTRACT

In mammals, the transcriptional activity of signal transducer and activator of transcription 3 (STAT3) is regulated by the deacetylase SIRT1. However, whether the newly described nongenomic actions of STAT3 toward mitochondrial oxidative phosphorylation are dependent on SIRT1 is unclear. In this study, Sirt1 gene knock-out murine embryonic fibroblast (MEF) cells were used to delineate the role of SIRT1 in the regulation of STAT3 mitochondrial function. Here, we show that STAT3 mRNA and protein levels and the accumulation of serine-phosphorylated STAT3 in mitochondria were increased significantly in Sirt1-KO cells as compared with wild-type MEFs. Various mitochondrial bioenergetic parameters, such as the oxygen consumption rate in cell cultures, enzyme activities of the electron transport chain complexes in isolated mitochondria, and production of ATP and lactate, indicated that Sirt1-KO cells exhibited higher mitochondrial respiration as compared with wild-type MEFs. Two independent approaches, including ectopic expression of SIRT1 and siRNA-mediated knockdown of STAT3, led to reduction in intracellular ATP levels and increased lactate production in Sirt1-KO cells that were approaching those of wild-type controls. Comparison of profiles of phospho-antibody array data indicated that the deletion of SirT1 was accompanied by constitutive activation of the pro-inflammatory NF-κB pathway, which is key for STAT3 induction and increased cellular respiration in Sirt1-KO cells. Thus, SIRT1 appears to be a functional regulator of NF-κB-dependent STAT3 expression that induces mitochondrial biogenesis. These results have implications for understanding the interplay between STAT3 and SIRT1 in pro-inflammatory conditions.


Subject(s)
Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Mitochondria/metabolism , Oxygen Consumption/physiology , STAT3 Transcription Factor/biosynthesis , Sirtuin 1/metabolism , Adenosine Triphosphate/biosynthesis , Adenosine Triphosphate/genetics , Animals , Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Lactic Acid/metabolism , Mice , Mitochondria/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Phosphorylation , Phosphorylation , STAT3 Transcription Factor/genetics , Sirtuin 1/genetics
13.
Cancer Lett ; 296(2): 225-32, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20444544

ABSTRACT

The tumor suppressor protein p53 plays a key role in regulation of negative cellular growth in response to EGCG. To further explore the role of p53 signaling and elucidate the molecular mechanism, we employed colon cancer HCT116 cell line and its derivatives in which a specific transcriptional target of p53 is knocked down by homologous recombination. Cells expressing p53 and p21 accumulate in G1 upon treatment with EGCG. In contrast, same cells lacking p21 traverse through the cell cycle and eventually undergo apoptosis as revealed by TUNEL staining. Treatment with EGCG leads to induction of p53, p21 and PUMA in p21 wild-type, and p53 and PUMA in p21(-/-) cells. Ablation of p53 by RNAi protects p21(-/-) cells, thus indicating a p53-dependent apoptosis by EGCG. Furthermore, analysis of cells lacking PUMA or Bax with or without p21 but with p53 reveals that all the cells expressing p53 and p21 survived after EGCG treatment. More interestingly, cells lacking both PUMA and p21 survived ECGC treatment whereas those lacking p21 and Bax did not. Taken together, our results present a novel concept wherein p21-dependent growth arrest pre-empts and protects cells from otherwise, in its absence, apoptosis which is mediated by activation of pro-apoptotic protein PUMA. Furthermore, we find that p53-dependent activation of PUMA in response to EGCG directly leads to apoptosis with out requiring Bax as is the case in response to agents that induce DNA damage. p21, thus can be used as a molecular switch for therapeutic intervention of colon cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis/drug effects , Catechin/analogs & derivatives , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HCT116 Cells/physiology , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/physiology , Catechin/pharmacology , Cell Cycle/drug effects , DNA Damage , Genes, p53/drug effects , HCT116 Cells/pathology , Humans , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Transfection , Tumor Suppressor Protein p53/metabolism
14.
Proc Natl Acad Sci U S A ; 105(17): 6314-9, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18434539

ABSTRACT

p53-dependent G(1) and G(2) cell cycle checkpoints are activated in response DNA damage that help to maintain genomic stability. p53 also helps to protect cells from damage that occurs during S phase, for example, when the cells are starved for DNA precursors or irradiated with a low dose of UV. p53 is activated in normal cells starved for pyrimidine nucleotides by treatment with N-(phosphonacetyl)-l-aspartate (PALA). The treated cells progress through a first S phase with kinetics similar to those of untreated cells. However, the DNA of the treated cells begins to become damaged rapidly, within 12 h, as revealed by a comet assay, which detects broken DNA, and by staining for phosphorylated histone H2AX, which accumulates at sites of DNA damage. Because the cells survive, the damage must be reversible, suggesting single-strand breaks or gaps as the most likely possibility. The transiently damaged DNA stimulates activation of ATR and CHK1, which in turn catalyze the phosphorylation and accumulation of p53. Although PALA-induced DNA damage occurs only in dividing cells, the p53 that is activated is only competent to transcribe genes such as p21 and macrophage inhibitory cytokine 1 (whose products regulate G(2) and G(1) or S phase checkpoints, respectively) after the cells have exited the S phase during which damage occurs. We propose that p53 is activated by stimulation of mismatch repair in response to the misincorporation of deoxynucleotides into newly synthesized DNA, long before the lack of pyrimidine nucleoside triphosphates causes the rate of DNA synthesis to slow appreciably.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA/biosynthesis , Nucleotides/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Aspartic Acid/analogs & derivatives , Aspartic Acid/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Line , Checkpoint Kinase 1 , DNA-Binding Proteins/metabolism , Humans , Models, Biological , Phosphonoacetic Acid/analogs & derivatives , Phosphonoacetic Acid/pharmacology , Phosphorylation/drug effects , Pyrimidines/metabolism , S Phase/drug effects , Tumor Suppressor Proteins/metabolism
15.
Cancer Res ; 67(12): 5617-21, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575126

ABSTRACT

Virtually all human cancers encounter disruption of the "p53 network." From a therapeutic point of view, it is important to devise strategies that eliminate cancer cells, which are often defective in functional p53 and protect p53-expressing normal cells. By comparing the response of a pair of isogenic cell lines, we identify a plant-derived compound, Concanavalin A (Con A), which differentially kills p53-null cells. Further, we find that p53 family member, p73, plays a critical role that is unmasked in the absence of p53. Con A treatment leads to induction of p73 and several others that are important mediators of apoptosis and act downstream, such as p21, Bax, Foxo1a, and Bim. Inactivation of p73 reverses the expression of these proteins and apoptosis. Inhibition of Akt activation sensitizes otherwise resistant cells. These observations thus reveal a novel role for p73 in the regulation of Akt-Foxo1a-Bim signaling and apoptosis especially when p53 is absent.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Concanavalin A/pharmacology , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Bcl-2-Like Protein 11 , Blotting, Western , Cell Line , Fibroblasts/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , In Situ Nick-End Labeling , Membrane Proteins/drug effects , Plant Lectins/pharmacology , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Protein p73 , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
16.
Proc Natl Acad Sci U S A ; 104(13): 5419-24, 2007 Mar 27.
Article in English | MEDLINE | ID: mdl-17369354

ABSTRACT

Green tea polyphenol, epigallocatechin-3-gallate (EGCG) differentially regulates the cellular growth of cancer cells in a p53-dependent manner through apoptosis and/or cell cycle arrest. In an effort to further elucidate the mechanism of differential growth regulation by EGCG, we have investigated the role of the tyrosine phosphatase, SHP-2. Comparing the responses of mouse embryonic fibroblasts (MEFs), expressing either WT or functionally inactive/truncated SHP-2, we find that inactivation of SHP-2 remarkably sensitizes cells to EGCG-mediated killing. MEFs lacking functional SHP-2 undergo massive apoptosis upon treatment with EGCG. By comparing gene expression profiles, we have identified a set of transcriptional targets of p53 that are differentially modulated in cells undergoing apoptosis. Western blot and real-time PCR analyses of a select group of genes further confirm that the expression is SHP-2-dependent. Similar observations were made in MEFs lacking p53, confirming that the expression of these "p53 target genes" is p53-independent. In addition, EGCG treatment induced the expression of p73 mRNA and protein in both cell types, but not p63. Inactivation of p73 in cells expressing nonfunctional SHP-2 markedly inhibited apoptosis and p53 target gene expression. Although phosphorylation of JNK is differentially regulated by SHP2, it was found to be dispensable for EGCG-induced apoptosis and p53 target gene expression. Our results have identified SHP-2 as a negative regulator of EGCG-induced-apoptosis and have identified a subset of p53 target genes whose expression is paradoxically not mediated by p53 but by one of its family members, p73.


Subject(s)
Apoptosis , Catechin/analogs & derivatives , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/chemistry , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/chemistry , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/metabolism , Animals , Catechin/pharmacology , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Mice , Oxidative Stress , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Tea , Transcriptional Activation , Tumor Protein p73
17.
J Exp Biol ; 209(Pt 14): 2774-84, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16809468

ABSTRACT

The biogenic amine octopamine (OA) is involved in the regulation of honey bee behavioral development; brain levels are higher in foragers than bees working in the hive, especially in the antennal lobes, and treatment causes precocious foraging. We measured brain mRNA and protein activity of tyramine beta-hydroxylase (T betah), an enzyme vital for OA synthesis, in order to begin testing the hypothesis that this enzyme is responsible for the rising levels of OA during honey bee behavioral development. Brain OA levels were greater in forager bees than in bees engaged in brood care, as in previous studies, but T betah activity was not correlated with bee behavior. T betah mRNA levels, however, did closely track OA levels during behavioral development, and T betah mRNA was localized to previously identified octopaminergic neurons in the bee brain. Our results show that the transcription of this neurotransmitter synthetic enzyme is associated with regulation of social behavior in honey bees, but other factors may be involved.


Subject(s)
Bees/enzymology , Behavior, Animal/physiology , Mixed Function Oxygenases/metabolism , Amino Acid Sequence , Animals , Brain/anatomy & histology , Brain/metabolism , Gene Expression Regulation, Enzymologic , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/genetics , Molecular Sequence Data , Octopamine/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL