Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
J Korean Med Sci ; 35(48): e371, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33316855

ABSTRACT

BACKGROUND: Cyclic AMP (cAMP) signaling is activated by various hormones and neurotransmitters and regulates numerous physiological phenomena, including energy metabolism, gene expression, and proliferation. cAMP signaling plays a role in the repair of DNA damage, but its specific function is inconsistent in the literature. The present study aimed to investigate the mechanism of the different roles of cAMP signaling in DNA repair by analyzing the cell-type differences in the modulation of DNA repair by cAMP signaling following γ-ray irradiation. METHODS: cAMP signaling was activated in human malignant melanoma cells (SK-MEL-2 and SK-MEL-28), human uterine cervical cancer cells (HeLa and SiHa) and human non-small cell lung cancer cells (H1299 and A549) by expressing a constitutively active mutant of the long-form stimulatory α subunit of GTP-binding protein or by treating with isoproterenol and prostaglandin E2 before γ-ray irradiation. DNA damage was quantitated by western blot analysis of γ-H2AX, and non-homologous end joining (NHEJ) was assessed by fluorescent reporter plasmid repair assay and immunofluorescence of microscopic foci of XRCC4 and DNA-ligase IV. RESULTS: cAMP signaling modulated DNA damage, apoptosis and the NHEJ repair following γ-ray irradiation differently depending upon the cell type. cAMP signaling regulated the phosphorylation of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) at Ser2056 and Thr2609 in cell-type-specific manners following γ-ray irradiation, an activity that was mediated by protein kinase A. CONCLUSION: cAMP signaling modulates the NHEJ repair of γ-ray-induced DNA damage in melanoma cells, uterine cervical cancer cells and lung cancer cells in a cell-type-specific manner, and the modulation is likely mediated by protein kinase A-dependent phosphorylation of DNA-PKcs. This study suggests that cell- and tissue-specific modulation of DNA damage repair by cAMP signaling may contribute to improve the therapeutic efficiency of radiation therapy.


Subject(s)
Cyclic AMP/metabolism , DNA Breaks, Double-Stranded/radiation effects , Gamma Rays , Signal Transduction/radiation effects , Apoptosis/radiation effects , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair/radiation effects , DNA-Activated Protein Kinase/metabolism , Humans , Phosphorylation/radiation effects
2.
J Biol Chem ; 290(15): 9604-13, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25713071

ABSTRACT

The cAMP signaling system regulates various cellular functions, including metabolism, gene expression, and death. Sirtuin 6 (SIRT6) removes acetyl groups from histones and regulates genomic stability and cell viability. We hypothesized that cAMP modulates SIRT6 activity to regulate apoptosis. Therefore, we examined the effects of cAMP signaling on SIRT6 expression and radiation-induced apoptosis in lung cancer cells. cAMP signaling in H1299 and A549 human non-small cell lung cancer cells was activated via the expression of constitutively active Gαs plus treatment with prostaglandin E2 (PGE2), isoproterenol, or forskolin. The expression of sirtuins and signaling molecules were analyzed by Western blotting. Activation of cAMP signaling reduced SIRT6 protein expression in lung cancer cells. cAMP signaling increased the ubiquitination of SIRT6 protein and promoted its degradation. Treatment with MG132 and inhibiting PKA with H89 or with a dominant-negative PKA abolished the cAMP-mediated reduction in SIRT6 levels. Treatment with PGE2 inhibited c-Raf activation by increasing inhibitory phosphorylation at Ser-259 in a PKA-dependent manner, thereby inhibiting downstream MEK-ERK signaling. Inhibiting ERK with inhibitors or with dominant-negative ERKs reduced SIRT6 expression, whereas activation of ERK by constitutively active MEK abolished the SIRT6-depleting effects of PGE2. cAMP signaling also augmented radiation-induced apoptosis in lung cancer cells. This effect was abolished by exogenous expression of SIRT6. It is concluded that cAMP signaling reduces SIRT6 expression by promoting its ubiquitin-proteasome-dependent degradation, a process mediated by the PKA-dependent inhibition of the Raf-MEK-ERK pathway. Reduced SIRT6 expression mediates the augmentation of radiation-induced apoptosis by cAMP signaling in lung cancer cells.


Subject(s)
Cyclic AMP/metabolism , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Sirtuins/metabolism , Apoptosis/radiation effects , Blotting, Western , Cell Line, Tumor , Dinoprostone/pharmacology , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoquinolines/pharmacology , Leupeptins/pharmacology , MAP Kinase Signaling System/drug effects , Models, Biological , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sirtuins/genetics , Sulfonamides/pharmacology , Ubiquitin/metabolism
3.
Biochem Biophys Res Commun ; 470(2): 336-342, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26792731

ABSTRACT

This study aimed to investigate the roles of autophagy and the ubiquitin-proteasome system in the degradation of histone deacetylase 8 (HDAC8) and to clarify the mechanism by which cAMP signaling regulates this degradation. cAMP signaling was activated by treating H1299 non-small cell lung cancer cells with isoproterenol or forskolin/3-isobutyl-1-methylxanthine, and HDAC8 expression was assessed by western blot analysis. The inhibition of autophagy and ubiquitin-proteasome-dependent degradation increased HDAC8 expression. cAMP signaling inhibited JNK activation, which decreased the phosphorylation of Bcl-2, thereby reducing autophagy, and the phosphorylation of Itch, thereby reducing ubiquitination. These results suggest that the HDAC8 protein is degraded via autophagy and the ubiquitin-proteasome system and that cAMP signaling increases HDAC8 protein levels by reducing JNK-mediated autophagy and ubiquitin-proteasome-dependent degradation of the HDAC8 protein in H1299 lung cancer cells.


Subject(s)
Autophagy , Cyclic AMP/metabolism , Histone Deacetylases/metabolism , Lung Neoplasms/metabolism , MAP Kinase Kinase 4/metabolism , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Signal Transduction , Up-Regulation
4.
Mol Cancer ; 13: 36, 2014 Feb 24.
Article in English | MEDLINE | ID: mdl-24568192

ABSTRACT

BACKGROUND: The ataxia-telangiectasia mutated (ATM) protein kinase plays a central role in coordinating the cellular response to radiation-induced DNA damage. cAMP signaling regulates various cellular responses including metabolism and gene expression. This study aimed to investigate the mechanism through which cAMP signaling regulates ATM activation and cellular responses to ionizing radiation in lung cancer cells. METHODS: Lung cancer cells were transfected with constitutively active stimulatory G protein (GαsQL), and irradiated with γ-rays. The phosphorylation of ATM and protein phosphatase 2A was analyzed by western blotting, and apoptosis was assessed by western blotting, flow cytometry, and TUNNEL staining. The promoter activity of NF-κB was determined by dual luciferase reporter assay. BALB/c mice were treated with forskolin to assess the effect in the lung tissue. RESULTS: Transient expression of GαsQL significantly inhibited radiation-induced ATM phosphorylation in H1299 human lung cancer cells. Treatment with okadaic acid or knock down of PP2A B56δ subunit abolished the inhibitory effect of Gαs on radiation-induced ATM phosphorylation. Expression of GαsQL increased phosphorylation of the B56δ and PP2A activity, and inhibition of PKA blocked Gαs-induced PP2A activation. GαsQL enhanced radiation-induced cleavage of caspase-3 and PARP and increased the number of early apoptotic cells. The radiation-induced apoptosis was increased by inhibition of NF-κB using PDTC or inhibition of ATM using KU55933 or siRNA against ATM. Pretreatment of BALB/c mice with forskolin stimulated phosphorylation of PP2A B56δ, inhibited the activation of ATM and NF-κB, and augmented radiation-induced apoptosis in the lung tissue. GαsQL expression decreased the nuclear levels of the p50 and p65 subunits and NF-κB-dependent activity after γ-ray irradiation in H1299 cells. Pretreatment with prostaglandin E2 or isoproterenol increased B56δ phosphorylation, decreased radiation-induced ATM phosphorylation and increased apoptosis. CONCLUSIONS: cAMP signaling inhibits radiation-induced ATM activation by PKA-dependent activation of PP2A, and this signaling mechanism augments radiation-induced apoptosis by reducing ATM-dependent activation of NF-κB in lung cancer cells.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cyclic AMP/metabolism , Lung Neoplasms/genetics , Protein Phosphatase 2/biosynthesis , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cyclic AMP/genetics , DNA Damage/radiation effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice , Phosphorylation/genetics , Phosphorylation/radiation effects , Protein Phosphatase 2/metabolism , Signal Transduction/genetics , Signal Transduction/radiation effects
5.
BMC Cell Biol ; 14: 4, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23339468

ABSTRACT

BACKGROUND: Cadmium(Cd), a heavy metal, which has a potent harmful effects, is a highly stress-inducible material that is robustly expressed following disruption of homeostasis in the endoplasmic reticulum (ER) (so-called ER stress). The mechanism Cd induced cell death of neuroblastoma cells complex, involving cellular signaling pathways as yet incompletely defined but, in part, involving the generation of reactive oxygen species (ROS). Several studies have correlated GADD153 expression with cell death, but a mechanistic link between GADD153 and apoptosis has never been demonstrated. RESULTS: SH-SY5Y cells were treated Cd led to increase in intracellular ROS levels. ROS generation is not consistent with intracellular [Ca2+]. The exposure of neuroblastoma cells to Cd led to increase in intracellular GADD153 and Bak levels in a doses and time dependent manner. The induction of these genes by Cd was attenuated by NAC. Cd-induced apoptosis is decreased in GADD153 knockdown cells compared with normal cells. The effect of GADD153 on the binding of C/EBP to the Bak promoters were analyzed ChIP assay. Basal constitutive GADD153 recruitment to the -3,398/-3,380 region of the Bak promoter is observed in SH-SY5Y cells. CONCLUSIONS: The exposure of SH-SY5Y cells to Cd led to increase in intracellular ROS levels in a doses and time dependent manner. The generation of ROS result in the induction of GADD153 is causative of cadmium-induced apoptosis. GADD153 regulates Bak expression by its binding to promoter region (between -3,398 and -3,380). Therefore, we conclude that GADD153 sensitizes cells to ROS through mechanisms that involve up-regulation of BAK and enhanced oxidant injury.


Subject(s)
Apoptosis/drug effects , Cadmium/toxicity , Neurons/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/metabolism , Acetylcysteine/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Cadmium/chemistry , Cell Line, Tumor , Free Radical Scavengers/pharmacology , Humans , Neurons/drug effects , Promoter Regions, Genetic , Protein Binding/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
6.
Biochem Biophys Res Commun ; 422(2): 256-62, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22575451

ABSTRACT

Cyclic AMP is involved in the regulation of metabolism, gene expression, cellular growth and proliferation. Recently, the cAMP signaling system was found to modulate DNA-damaging agent-induced apoptosis by regulating the expression of Bcl-2 family proteins and inhibitors of apoptosis. Thus, we hypothesized that the cAMP signaling may modulate DNA repair activity, and we investigated the effects of the cAMP signaling system on γ-ray-induced DNA damage repair in lung cancer cells. Transient expression of a constitutively active mutant of stimulatory G protein (GαsQL) or treatment with forskolin, an adenylyl cyclase activator, augmented radiation-induced DNA damage and inhibited repair of the damage in H1299 lung cancer cells. Expression of GαsQL or treatment with forskolin or isoproterenol inhibited the radiation-induced expression of the XRCC1 protein, and exogenous expression of XRCC1 abolished the DNA repair-inhibiting effect of forskolin. Forskolin treatment promoted the ubiquitin and proteasome-dependent degradation of the XRCC1 protein, resulting in a significant decrease in the half-life of the protein after γ-ray irradiation. The effect of forskolin on XRCC1 expression was not inhibited by PKA inhibitor, but 8-pCPT-2'-O-Me-cAMP, an Epac-selective cAMP analog, increased ubiquitination of XRCC1 protein and decreased XRCC1 expression. Knockdown of Epac1 abolished the effect of 8-pCPT-2'-O-Me-cAMP and restored XRCC1 protein level following γ-ray irradiation. From these results, we conclude that the cAMP signaling system inhibits the repair of γ-ray-induced DNA damage by promoting the ubiquitin-proteasome dependent degradation of XRCC1 in an Epac-dependent pathway in lung cancer cells.


Subject(s)
Cyclic AMP/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Cell Line, Tumor , Gamma Rays , Humans , Proteolysis , Ubiquitin/metabolism , X-ray Repair Cross Complementing Protein 1
7.
Ann Surg Oncol ; 19(11): 3441-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22618722

ABSTRACT

BACKGROUND: The prognostic significance of microsatellite instability (MSI) in colorectal cancers (CRCs) has been addressed in many studies since the initial description of better survival rates in MSI-positive (MSI+) tumors than in MSI-negative (MSI-) tumors. Recent studies have demonstrated that a higher degree of hypomethylation of long interspersed nuclear element-1 (L1) is related to poor prognosis of CRCs and that a wide variation of L1 methylation levels exist within MSI+ CRCs. Our aim was to identify whether L1 and Alu methylation status could predict clinical outcomes within MSI+ CRCs. METHODS: We analyzed 207 MSI+ CRCs for their methylation levels in L1 and Alu repetitive DNA elements using pyrosequencing and correlated them with clinicopathological information including survival data. RESULTS: Univariate survival analysis showed that low Alu methylation status (<18.60%) and low L1 methylation status (<53.00%) were significantly associated with shorter overall survival time (log-rank test, P = 0.009 and P < 0.001, respectively). Multivariate analysis using nine parameters (Alu methylation status, L1 methylation status, patient's age, disease stage [tumor, node, metastasis staging system], differentiation, Crohn-like lymphoid reaction, KRAS/BRAF mutation status, CpG island methylator phenotype [CIMP] status, and peritumoral lymphocytic infiltration), which were significantly prognostic in MSI+ CRCs, revealed that low L1 methylation status was an independent prognostic factor of MSI+ CRCs (P = 0.009), whereas low Alu methylation status was not. CONCLUSIONS: Clinical outcomes of MSI+ CRCs depend on L1 methylation status, suggesting that lower L1 methylation status serves as a significant prognostic parameter of adverse prognosis in MSI+ CRCs.


Subject(s)
Alu Elements , Carcinoma/genetics , Carcinoma/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Methylation , Long Interspersed Nucleotide Elements , DNA Mutational Analysis , Female , Humans , Kaplan-Meier Estimate , Male , Microsatellite Instability , Middle Aged , Multivariate Analysis , Neoplasm Staging , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Statistics, Nonparametric , ras Proteins/genetics
8.
Cancer Sci ; 102(4): 837-44, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21255191

ABSTRACT

Treatment with cisplatin (cis-dichlorodiammineplatinum (II)) induces DNA double-stranded breaks and apoptosis in many human cancer cells. We have reported that heterotrimeric stimulatory GTP-binding proteins (Gαs) can modulate the apoptotic response of several cancer cells. This study investigated the effect of Gαs on apoptosis triggered by cisplatin and its underlying molecular mechanism in cervical cancer cells. Stable expression of constitutively active Gαs (GαsQL) decreased the release of cytochrome c from the mitochondria to the cytosol and cleavage of caspase-3 and poly(ADP-ribose) polymerases in HeLa cells treated with 30 µM cisplatin, indicating that Gαs inhibited cisplatin-induced apoptosis. Treatment with forskolin also inhibited apoptosis of C33A and CaSKi cervical cancer cells. Expression of GαsQL increased the expression of the X-linked inhibitor of apoptosis protein (XIAP) and partially maintained increased XIAP after cisplatin treatment. Knockdown of XIAP by siRNA augmented apoptosis. Expression of GαsQL increased XIAP mRNA; this increase was inhibited by a protein kinase A inhibitor and cAMP response element (CRE) decoy. A cAMP response element (CRE)-like element at -1396 bp in the XIAP promoter was found to mediate the induction of XIAP by Gαs. In addition, expression of GαsQL protected against the ubiquitin/proteasome-dependent degradation of the XIAP protein. This study shows that Gαs inhibits cisplatin-induced apoptosis by increasing transcription of XIAP and by decreasing degradation of XIAP protein in HeLa cervical cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , GTP-Binding Protein alpha Subunits, Gs/metabolism , Uterine Cervical Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Blotting, Western , Female , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Luciferases/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
9.
Carcinogenesis ; 31(3): 402-10, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20015862

ABSTRACT

To investigate the mechanism by which the human papillomavirus (HPV) E5 protein contributes to the carcinogenesis of uterine cervical cancer, we studied the effect of HPV E5 on apoptosis of cervical cancer cells and its underlying mechanism. Expression of HPV16 E5 protein inhibited hydrogen peroxide-induced apoptosis in C-33A cervical cancer cells. E5 decreased the expression of Bax protein, and exogenous expression of Bax abolished the anti-apoptotic effect of E5. Knockdown of E5 by small interfering RNA sensitized CaSki cervical cancer cells to hydrogen peroxide-induced apoptosis with concurrent increase in Bax expression. Transient expression of E5 significantly increased the degradation rate of Bax protein by inducing the ubiquitination. The E5-induced decrease in Bax expression was inhibited by a cyclooxygenase-2 (COX-2) inhibitor, prostaglandin E2 (PGE(2)) receptor antagonists and cyclic adenosine monophosphate-dependent protein kinase (PKA) inhibitor. Treatment with PGE(2) decreased the expression of Bax and inhibited hydrogen peroxide-induced apoptosis of C-33A cells. We concluded that HPV16 E5 protein inhibits hydrogen peroxide-induced apoptosis of cervical cancer cells by stimulating the ubiquitin-proteasome-mediated degradation of Bax protein, and the pathway involves COX-2, PGE(2) and PKA. This finding suggests the possibility that HPV 16 E5 protein contributes to cervical carcinogenesis by inhibiting apoptosis of transformed cervical epithelial cells.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Neoplasm Proteins/physiology , Oncogene Proteins, Viral/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/physiology , Uterine Cervical Neoplasms/pathology , bcl-2-Associated X Protein/metabolism , Androstadienes/pharmacology , Biphenyl Compounds/pharmacology , Carcinoma, Squamous Cell/metabolism , Cell Line, Transformed/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Cycloheximide/pharmacology , Cyclooxygenase 2/physiology , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/pharmacology , Female , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Isoquinolines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Sulfonamides/pharmacology , Uterine Cervical Neoplasms/metabolism , Wortmannin , Xanthones/pharmacology
10.
Biochem Biophys Res Commun ; 393(2): 190-5, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20117092

ABSTRACT

Ultraviolet (UV) radiation induces cyclooxygenase-2 expression to produce cellular responses including aging and carcinogenesis in skin. We hypothesised that heterotrimeric G proteins mediate UV-induced COX-2 expression by stimulating secretion of soluble HB-EGF (sHB-EGF). In this study, we aimed to elucidate the role and underlying mechanism of the alpha subunit of Gq protein (Galphaq) in UVB-induced HB-EGF secretion and COX-2 induction. We found that expression of constitutively active Galphaq (GalphaqQL) augmented UVB-induced HB-EGF secretion, which was abolished by knockdown of Galphaq with shRNA in HaCaT human keratinocytes. Galphaq was found to mediate the UVB-induced HB-EGF secretion by sequential activation of phospholipase C (PLC), protein kinase Cdelta (PKCdelta), and matrix metaloprotease-2 (MMP-2). Moreover, GalphaqQL mediated UVB-induced COX-2 expression in an HB-EGF-, EGFR-, and p38-dependent manner. From these results, we concluded that Galphaq mediates UV-induced COX-2 expression through activation of EGFR by HB-EGF, of which ectodomain shedding was stimulated through sequential activation of PLC, PKCdelta and MMP-2 in HaCaT cells.


Subject(s)
Cyclooxygenase 2/biosynthesis , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Keratinocytes/radiation effects , Ultraviolet Rays , Cell Transformation, Neoplastic/metabolism , Enzyme Activation , ErbB Receptors/agonists , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Keratinocytes/enzymology , Matrix Metalloproteinase 2/metabolism , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/metabolism , Protein Kinase C beta , Skin Aging , Skin Neoplasms/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Sci Rep ; 10(1): 14455, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32879366

ABSTRACT

DNA double-strand breaks (DSB) are formed by various exogenous and endogenous factors and are repaired by homologous recombination and non-homologous end joining (NHEJ). DNA-dependent protein kinase (DNA-PK) is the principal enzyme for NHEJ. We explored the role and the underlying mechanism of cAMP signaling in the NHEJ repair of DSBs resulted from gamma ray irradiation to non-small cell lung cancer (NSLC) cells. Activated cAMP signaling by expression of an activated stimulatory GTP-binding protein or by pretreatment with isoproterenol and prostaglandin E2, delayed the repair of DSBs resulted from gamma ray irradiation, and the delaying effects depended on protein kinase A (PKA). Activated cAMP signaling suppressed XRCC4 and DNA ligase IV recruitment into DSB foci, and reduced phosphorylation at T2609 in DNA-PK catalytic subunit (DNA-PKcs) with a concomitant increase in phosphorylation at S2056 in PKA-dependent ways following gamma ray irradiation. cAMP signaling decreased phosphorylation of T2609 by protein phosphatase 2A-dependent inhibition of ATM. We conclude that cAMP signaling delays the repair of gamma ray-induced DNA DSBs in NSLC cells by inhibiting NHEJ via PKA-dependent pathways, and that cAMP signaling differentially modulates DNA-PKcs phosphorylation at S2056 and T2609, which might contribute to the inhibition of NHEJ in NSLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , DNA End-Joining Repair/genetics , DNA Ligase ATP/genetics , DNA-Binding Proteins/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Catalytic Domain/genetics , Cyclic AMP/genetics , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA End-Joining Repair/radiation effects , DNA Repair/radiation effects , DNA-Activated Protein Kinase/genetics , Gamma Rays/adverse effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Phosphorylation/radiation effects , Signal Transduction/radiation effects
12.
Carcinogenesis ; 30(5): 753-7, 2009 May.
Article in English | MEDLINE | ID: mdl-19321801

ABSTRACT

The human papillomavirus (HPV) E6 and E7 oncoproteins play important roles in cervical carcinogenesis through multiple mechanisms, including upregulation of cyclooxygenase-2 (COX-2), which has been shown to be involved in both carcinogenesis and cancer progression. To explore the role of E5 in cervical carcinogenesis, we herein investigated the effect of HPV 16 E5 on COX-2 expression. Our results revealed that E5 induced COX-2 expression through the epidermal growth factor receptor-signaling pathway, with nuclear factor-kappaB (NF-kappaB) and activator protein-1 (AP-1) acting as critical factors in E5-induced COX-2 expression. NF-kappaB inhibition blocked COX-2 expression more potently than inhibition of AP-1. Our findings collectively suggest that the HPV 16 E5 oncoprotein mediates cervical carcinogenesis at least in part via upregulation of COX-2 expression through NF-kappaB and AP-1, with NF-kappaB playing a larger role.


Subject(s)
Cyclooxygenase 2/genetics , NF-kappa B/physiology , Oncogene Proteins, Viral/physiology , Transcription Factor AP-1/physiology , Cell Line , Cell Line, Tumor , Culture Media , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Human papillomavirus 16/physiology , Humans , Kidney/embryology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/virology
13.
Carcinogenesis ; 30(1): 141-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18849297

ABSTRACT

Human papillomavirus (HPV) is the major cause of uterine cervical cancer, but the role of the HPV E5 in carcinogenesis is not clearly understood. Prostaglandins are known to contribute to carcinogenesis of cervical cancer, and we therefore investigated the effect of HPV16 E5 on the expression of prostaglandin E2 (PGE2) receptors and underlying mechanisms. Stable expression of the E5 induced expression of the EP4 subtype of PGE2 receptors in C33A cervical cancer cells, and transfection of E5 small interfering RNA (siRNA) decreased it. EP4 protein expression was increased in human cervical cancer tissues, and EP4 mediated E5-induced increase in anchorage-independent colony formation and vascular endothelial growth factor expression. E5 induced cyclooxygenase-2 (COX-2) expression, and COX-2 increased PGE2 secretion and EP4 expression. The induction of EP4 by PGE2 and E5 was inhibited by an EP4 antagonist, inhibitors of cyclic adenosine monophosphate-dependent protein kinase or phosphatidylinositol 3-kinase, and a cyclic adenosine monophosphate response element (CRE) decoy. E5 increased the luciferase expression controlled by a variant CRE of the EP4 promoter, and it also increased the binding of cyclic adenosine monophosphate response element binding protein (CREB) to oligonucleotides containing this CRE. We conclude that the HPV16 E5 protein induces EP4 receptor protein in cervical cancer cells and that this induction involves epidermal growth factor receptor, COX-2, PGE2, EP2 and EP4, protein kinase A, CREB and CRE.


Subject(s)
Cyclic AMP/metabolism , Oncogene Proteins, Viral/physiology , Receptors, Prostaglandin E/genetics , Uterine Cervical Neoplasms/metabolism , Base Sequence , Cell Line, Tumor , Cyclic AMP/physiology , Cyclic AMP Response Element-Binding Protein/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclooxygenase 2/metabolism , DNA Primers , Dinoprostone/physiology , Electrophoretic Mobility Shift Assay , ErbB Receptors/metabolism , Female , Humans , Immunohistochemistry , Luminescence , Promoter Regions, Genetic , RNA Interference , Receptors, Prostaglandin E, EP4 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/pathology
14.
J Neurochem ; 111(2): 344-54, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19659461

ABSTRACT

This study examined the role of calcineurin, a major calcium-dependent protein phosphatase, in dephosphorylating Ser-9 and activating glycogen synthase kinase-3beta (GSK-3beta). Treatment with calcineurin inhibitors increased phosphorylation of GSK-3beta at Ser-9 in SH-SY5Y human neuroblastoma cells. The over-expression of a constitutively active calcineurin mutant, calcineurin A beta (1-401), led to a significant decrease in phosphorylation at Ser-9, an increase in the activity of GSK-3beta, and an increase in the phosphorylation of tau. K(m) of calcineurin for a GSK-3beta phosphopeptide was 469.3 microM, and specific activity of calcineurin was 15.2 nmol/min/mg. In addition, calcineurin and GSK-3beta were co-immunoprecipitated in neuron-derived cells and brain tissues, and calcineurin formed a complex only with dephosphorylated GSK-3beta. We conclude that in vitro, calcineurin can dephosphorylate GSK-3beta at Ser-9 and form a stable complex with GSK-3beta, suggesting the possibility that calcineurin regulates the dephosphorylation and activation of GSK-3betain vivo.


Subject(s)
Calcineurin/genetics , Calcineurin/metabolism , Glycogen Synthase Kinase 3/metabolism , Neurons/enzymology , Animals , Calcineurin Inhibitors , Cell Line, Tumor , Cyclosporine/pharmacology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Glioma , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice , Mutation , Neuroblastoma , Neurons/cytology , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Tacrolimus/pharmacology , Transfection
15.
Cancer Sci ; 100(6): 1069-74, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19320642

ABSTRACT

The present study aimed to investigate the effect of the stimulatory heterotrimeric GTP-binding (Gs) protein signaling system on cisplatin-induced apoptosis of lung cancer cells and its underlying mechanism as an attempt to develop a novel strategy to improve the therapeutic efficacy of cisplatin. Overexpression of the constitutively active alpha subunit of Gs (GalphasQL) in A549 human lung cancer cells increased cisplatin-induced apoptosis, and knockdown of Galphas with small hairpin RNA decreased the percentage of apoptotic cells. GalphasQL increased the expression of the proapoptotic proteins B-cell leukemia/lymphoma-2 genes (Bcl-2) homologous antagonist killer protein (Bak) and Bcl-2 associated X protein (Bax), and decreased the expression of the antiapoptotic proteins Bcl-2 and Bcl-Xlong protein. Knockdown of Bak blocked the augmentative effects of GalphasQL. GalphasQL decreased the degradation rate of the Bak protein, and increased Bak mRNA transcript levels. GalphasQL increased Bak-luciferase activity in a protein kinase A and cyclic AMP response element-dependent manner. GalphasQL also augmented cisplatin-induced apoptosis of H1299 human lung cancer cells that lack functional p53. From this study, it is concluded that Galphas augments cisplatin-induced apoptosis of lung cancer cells partially through upregulating Bak expression by increasing transcription and by decreasing the rate of protein degradation.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Heterotrimeric GTP-Binding Proteins/metabolism , Lung Neoplasms/pathology , bcl-2 Homologous Antagonist-Killer Protein/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Primers , Flow Cytometry , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Transfection
16.
Biochem Biophys Res Commun ; 381(2): 153-8, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19233129

ABSTRACT

Inhibitory heterotrimeric GTP-binding proteins (Gi proteins) mediate a variety of signaling pathways by coupling receptors and effectors to regulate cellular proliferation, differentiation, and apoptosis. However, the role of Gi proteins in the modulation of hydrogen peroxide-induced apoptosis is not clearly understood. Thus, we investigated the effect of Gi proteins on hydrogen peroxide-induced apoptosis and the underlying mechanisms in H1299 human lung cancer cells. The stable expression of constitutively active alpha subunits of Gi1 (Galphai1QL), Gi2, or Gi3 inhibited hydrogen peroxide-induced apoptosis. The expression of Galphai1QL up-regulated Bcl-2 expression, and the knockdown of Bcl-2 with siRNA abolished the anti-apoptotic effect of Galphai1QL. Galphai1 induced the transcription of Bcl-2 by activation of NF-kappaB, which resulted from an increase in NF-kappaB p50 protein. We conclude that Galphai1 inhibits hydrogen peroxide-induced apoptosis of H1299 lung cancer cells by up-regulating the transcription of Bcl-2 through a p50-mediated NF-kappaB activation.


Subject(s)
Apoptosis , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Lung Neoplasms/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line, Tumor , Humans , Hydrogen Peroxide/pharmacology , Up-Regulation
17.
Psychiatry Res ; 167(1-2): 80-7, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19342105

ABSTRACT

Repeated administration of NMDA antagonists can induce behavioral alterations that mimic symptoms of psychosis, as seen in schizophrenia. JNK, one of the MAPKs, and c-Jun, its downstream target molecule, play important roles in regulating apoptosis in neural cells, and have been suggested as being associated with the pathophysiology of psychosis and the mechanism of action of some antipsychotics. We investigated changes in the JNK-c-Jun pathway and other Jun family proteins in the rat frontal cortex after single and repeated administration of MK-801 to examine acute and chronic responses. Neither the protein level nor the phosphorylation of JNK changed after single or repeated doses of MK-801. However, after repeated treatments, but not a single treatment, with MK-801, a down-regulation occurred in the protein level and of Ser73 phosphorylation of c-Jun in the rat frontal cortex. Other members of the Jun family, JunB and JunD, were unchanged. Repeated exposure to MK-801 down-regulated the phosphorylation and protein level of c-Jun in the rat frontal cortex, which may be related to the long-term effects of chronic treatment with MK-801.


Subject(s)
Dizocilpine Maleate/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Frontal Lobe/metabolism , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Antipsychotic Agents/pharmacology , Apoptosis/drug effects , Behavior, Animal/drug effects , Down-Regulation/drug effects , Frontal Lobe/drug effects , Male , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/chemically induced , Schizophrenia/metabolism , Signal Transduction/drug effects
18.
Cancer Sci ; 99(6): 1117-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18384434

ABSTRACT

The role of nuclear factor-kappaB (NF-kappaB) activation in cancer cell apoptosis appears to be tailored specifically for each cell type and the type of NF-kappaB inducer. The present study aimed to determine whether or not NF-kappaB activation is associated with chemosensitivity to doxorubicin (DOX) using the DOX-sensitive SNU-601 and DOX-resistant SNU-216 gastric cancer cell lines. The effect of NF-kappaB activation on DOX (1 microg/mL) sensitivity was analyzed after the suppression of NF-kappaB activation using transfection of the super-suppressive mutant form of IkappaBalpha (mIkappaBalpha) or pretreatment with pyrrolidine dithiocarbamate. In addition, the association between NF-kappaB and manganese superoxide dismutase (MnSOD) in relation to DOX sensitivity was analyzed after the modulation of MnSOD expression. The NF-kappaB activity was much higher in DOX-resistant SNU-216 cells than in DOX-sensitive SNU-601 cells before and after DOX treatment. Overexpression of mIkappaBalpha or pyrrolidine dithiocarbamate pretreatment decreased the DOX resistance in SNU-601 cells with low MnSOD expression, but not in SNU-216 cells with high MnSOD expression. In comparison, the overexpression of MnSOD, which also suppressed NF-kappaB activation in both cell lines, increased DOX resistance in SNU-601 cells. Blocking of MnSOD expression using RNA interference techniques increased DOX sensitivity in SNU-216 cells, which was further augmented by the additional inhibition of NF-kappaB activity. Our results showed that whether NF-kappaB contributes to DOX sensitivity in gastric cancer cells is determined by the level of MnSOD expression. Thus, targeting both MnSOD and NF-kappaB may be helpful for increasing the efficacy of DOX treatment of DOX-resistant SNU gastric cancer cells.


Subject(s)
Doxorubicin/pharmacology , Drug Resistance, Neoplasm , NF-kappa B/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Superoxide Dismutase/metabolism , Blotting, Western , Cell Nucleus/metabolism , Electrophoretic Mobility Shift Assay , Humans , Hydrogen Peroxide/metabolism , I-kappa B Proteins/metabolism , Luciferases/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Pyrrolidines/pharmacology , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Stomach Neoplasms/pathology , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Thiocarbamates/pharmacology , Tumor Cells, Cultured
19.
Exp Mol Med ; 40(4): 435-44, 2008 Aug 31.
Article in English | MEDLINE | ID: mdl-18779656

ABSTRACT

Repeated electroconvulsive seizure (ECS), a model for electroconvulsive therapy (ECT), exerts neuroprotective and proliferative effects in the brain. This trophic action of ECS requires inhibition of apoptotic activity, in addition to activation of survival signals. c-Myc plays an important role in apoptosis of neurons, in cooperation with the Bcl-2 family proteins, and its activity and stability are regulated by phosphorylation and ubiquitination. We examined c-Myc and related proteins responsible for apoptosis after repeated ECS. In the rat frontal cortex, repeated ECS for 10 days reduced the total amount of c-Myc, while increasing phosphorylation of c-Myc at Thr58, which reportedly induces degradation of c-Myc. As expected, ubiquitination of both phosphorylated and total c-Myc increased after 10 days ECS, suggesting that ECS may reduce c-Myc protein level via ubiquitination-proteasomal degradation. Bcl-2 family proteins, caspase, and poly(ADP-ribose) polymerase (PARP) were investigated to determine the consequence of down-regulating c-Myc. Protein levels of Bcl-2, Bcl-X(L), Bax, and Bad showed no change, and cleavage of caspase-3 and PARP were not induced. However, phosphorylation of Bad at Ser-155 and binding of Bad to 14-3-3 increased without binding to Bcl-X(L) after repeated ECS, implying that repeated ECS sequesters apoptotic Bad and frees pro-survival Bcl-XL. Taken together, c-Myc down-regulation via ubiquitination-proteasomal degradation and Bad inactivation by binding to 14-3-3 may be anti-apoptotic mechanisms elicited by repeated ECS in the rat frontal cortex. This finding further supports the trophic effect of ECS blocking apoptosis as a possible therapeutic effect of ECT.


Subject(s)
Electroconvulsive Therapy/adverse effects , Frontal Lobe/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Seizures/metabolism , bcl-Associated Death Protein/metabolism , 14-3-3 Proteins/metabolism , Animals , Down-Regulation , Male , Models, Biological , Neurons/metabolism , Periodicity , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Seizures/etiology , Tumor Cells, Cultured , Ubiquitination , bcl-Associated Death Protein/antagonists & inhibitors
20.
Exp Mol Med ; 40(5): 479-85, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18985005

ABSTRACT

Hypoxia-inducible factor 1alpha (HIF-1alpha) is rapidly degraded by the ubiquitin-proteasome pathway under normoxic conditions. Ubiquitination of HIF-1alpha is mediated by interaction with von Hippel-Lindau tumor suppressor protein (pVHL). In our previous report, we found that hypoxia-induced active signal transducer and activator of transcription3 (STAT3) accelerated the accumulation of HIF-1alpha protein and prolonged its half-life in solid tumor cells. However, its specific mechanisms are not fully understood. Thus, we examined the role of STAT3 in the mechanism of pVHL-mediated HIF-1alpha stability. We found that STAT3 interacts with C-terminal domain of HIF-1alpha and stabilizes HIF-1alpha by inhibition of pVHL binding to HIF-1alpha. The binding between HIF-1alpha and pVHL, negative regulator of HIF-1alpha stability, was interfered dose-dependently by overexpressed constitutive active STAT3. Moreover, we found that the enhanced HIF-1alpha protein levels by active STAT3 are due to decrease of poly-ubiquitination of HIF-1alpha protein via inhibition of interaction between pVHL and HIF-1alpha. Taken together, our results suggest that STAT3 decreases the pVHL-mediated ubiquitination of HIF-1alpha through competition with pVHL for binding to HIF-1 alpha, and then stabilizes HIF-1alpha protein levels.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , STAT3 Transcription Factor/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Immunoprecipitation , Protein Binding , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transfection , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL