Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 15(11): 1503-1516, 2017 11.
Article in English | MEDLINE | ID: mdl-28801308

ABSTRACT

EGFR signaling has been implicated in hypoxia-associated resistance to radiation or chemotherapy. Non-small cell lung carcinomas (NSCLC) with activating L858R or ΔE746-E750 EGFR mutations exhibit elevated EGFR activity and downstream signaling. Here, relative to wild-type (WT) EGFR, mutant (MT) EGFR expression significantly increases radiosensitivity in hypoxic cells. Gene expression profiling in human bronchial epithelial cells (HBEC) revealed that MT-EGFR expression elevated transcripts related to cell cycle and replication in aerobic and hypoxic conditions and downregulated RAD50, a critical component of nonhomologous end joining and homologous recombination DNA repair pathways. NSCLCs and HBEC with MT-EGFR revealed elevated basal and hypoxia-induced γ-H2AX-associated DNA lesions that were coincident with replication protein A in the S-phase nuclei. DNA fiber analysis showed that, relative to WT-EGFR, MT-EGFR NSCLCs harbored significantly higher levels of stalled replication forks and decreased fork velocities in aerobic and hypoxic conditions. EGFR blockade by cetuximab significantly increased radiosensitivity in hypoxic cells, recapitulating MT-EGFR expression and closely resembling synthetic lethality of PARP inhibition.Implications: This study demonstrates that within an altered DNA damage response of hypoxic NSCLC cells, mutant EGFR expression, or EGFR blockade by cetuximab exerts a synthetic lethality effect and significantly compromises radiation resistance in hypoxic tumor cells. Mol Cancer Res; 15(11); 1503-16. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Replication , DNA/metabolism , ErbB Receptors/genetics , Lung Neoplasms/genetics , A549 Cells , Acid Anhydride Hydrolases , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cetuximab/pharmacology , DNA Damage , DNA Repair , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Humans , Lung Neoplasms/drug therapy , Mutation , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology
2.
Sci Rep ; 5: 11305, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26065573

ABSTRACT

Carbon-ion radiotherapy (CIRT) holds promise to treat inoperable locally-advanced non-small cell lung carcinoma (NSCLC), a disease poorly controlled by standard chemoradiotherapy using X-rays. Since CIRT is an extremely limited medical resource, selection of NSCLC patients likely to benefit from it is important; however, biological predictors of response to CIRT are ill-defined. The present study investigated the association between the mutational status of EGFR and KRAS, driver genes frequently mutated in NSCLC, and the relative biological effectiveness (RBE) of carbon-ion beams over X-rays. The assessment of 15 NSCLC lines of different EGFR/KRAS mutational status and that of isogenic NSCLC lines expressing wild-type or mutant EGFR revealed that EGFR-mutant NSCLC cells, but not KRAS-mutant cells, show low RBE. This was attributable to (i) the high X-ray sensitivity of EGFR-mutant cells, since EGFR mutation is associated with a defect in non-homologous end joining, a major pathway for DNA double-strand break (DSB) repair, and (ii) the strong cell-killing effect of carbon-ion beams due to poor repair of carbon-ion beam-induced DSBs regardless of EGFR mutation status. These data highlight the potential of EGFR mutation status as a predictor of response to CIRT, i.e., CIRT may show a high therapeutic index in EGFR mutation-negative NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line, Tumor , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Heavy Ion Radiotherapy , Humans , Lung Neoplasms/radiotherapy , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
3.
Int J Oncol ; 46(3): 989-98, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25573239

ABSTRACT

Although cytotoxic chemotherapy is essential in epidermal growth factor receptor (EGFR)­mutated non­small cell lung cancer (NSCLC), it is unclear which regimen is most effective. We retrospectively compared the efficacy of standard platinum­based chemotherapy with that of combination chemotherapy using vinorelbine (VNR) plus dihydropyrimidine dehydrogenase­inhibitory fluoropyrimidine (DIF) in EGFR­mutated lung adenocarcinomas, and we investigated a potential mechanism by which the combination chemotherapy of VNR + DIF was favorable in the treatment of EGFR­mutated lung adenocarcinoma in vitro. In our retrospective analysis, the response rate and disease control rate afforded by the VNR + DIF treatment tended to be better than those by platinum­based chemotherapy, and the progression­free survival of the 24 VNR + DIF­treated patients was significantly longer than that of the 15 platinum­based chemotherapy patients. In EGFR­mutated PC9 cells, VNR induced EGFR dephosphorylation at a clinically achievable concentration. 1BR3­LR cells, a line of fibroblast cells transfected with a mutant EGFR construct, were completely resistant to gefitinib in the medium containing 10% fetal bovine serum (FBS), whereas the sensitivity of these cells to gefitinib was increased in 0.5% FBS­containing medium. Similarly, the sensitivity of 1BR3­LR cells to VNR was increased when they were cultured in low­serum condition. In addition, sodium orthovanadate (Na3VO4) inhibited the EGFR dephosphorylation induced by VNR or gefitinib and suppressed the cell growth inhibition by these agents in PC9 cells. VNR and gefitinib showed synergistic cell growth inhibition in combination with 5­fluorouracil (5­FU) in PC9 cells. We propose that the EGFR dephosphorylation induced by VNR is related to cell growth inhibitory activity of VNR, and that this is one of the mechanisms of the synergistic effect of VNR + 5­FU in EGFR­mutated lung cancer cells. In conclusion, the combination chemotherapy of VNR + DIF may be a promising treatment for NSCLC patients with EGFR mutations.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor/drug effects , Dihydrouracil Dehydrogenase (NADP)/antagonists & inhibitors , Drug Combinations , ErbB Receptors/metabolism , Female , Fluorouracil/administration & dosage , Gefitinib , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Oxonic Acid/administration & dosage , Quinazolines/pharmacology , Retrospective Studies , Tegafur/administration & dosage , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/pharmacology , Vinorelbine
4.
J Biol Chem ; 288(29): 21197-21207, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23740244

ABSTRACT

Recently, we have shown that CXCL12/CXCR4 signaling plays an important role in gemcitabine resistance of pancreatic cancer (PC) cells. Here, we explored the effect of gemcitabine on this resistance mechanism. Our data demonstrate that gemcitabine induces CXCR4 expression in two PC cell lines (MiaPaCa and Colo357) in a dose- and time-dependent manner. Gemcitabine-induced CXCR4 expression is dependent on reactive oxygen species (ROS) generation because it is abrogated by pretreatment of PC cells with the free radical scavenger N-acetyl-L-cysteine. CXCR4 up-regulation by gemcitabine correlates with time-dependent accumulation of NF-κB and HIF-1α in the nucleus. Enhanced binding of NF-κB and HIF-1α to the CXCR4 promoter is observed in gemcitabine-treated PC cells, whereas their silencing by RNA interference causes suppression of gemcitabine-induced CXCR4 expression. ROS induction upon gemcitabine treatment precedes the nuclear accumulation of NF-κB and HIF-1α, and suppression of ROS diminishes these effects. The effect of ROS on NF-κB and HIF-1α is mediated through activation of ERK1/2 and Akt, and their pharmacological inhibition also suppresses gemcitabine-induced CXCR4 up-regulation. Interestingly, our data demonstrate that nuclear accumulation of NF-κB results from phosphorylation-induced degradation of IκBα, whereas HIF-1α up-regulation is NF-κB-dependent. Lastly, our data demonstrate that gemcitabine-treated PC cells are more motile and exhibit significantly greater invasiveness against a CXCL12 gradient. Together, these findings reinforce the role of CXCL12/CXCR4 signaling in gemcitabine resistance and point toward an unintended and undesired effect of chemotherapy.


Subject(s)
Deoxycytidine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Up-Regulation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Deoxycytidine/adverse effects , Deoxycytidine/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects , Up-Regulation/genetics , Gemcitabine
5.
Mol Cancer Res ; 10(10): 1359-68, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22923485

ABSTRACT

The EGF receptor (EGFR) contributes to tumor radioresistance, in part, through interactions with the catalytic subunit of DNA-dependent protein kinase (DNA-PKc), a key enzyme in the nonhomologous end joining DNA repair pathway. We previously showed that EGFR-DNA-PKcs interactions are significantly compromised in the context of activating mutations in EGFR in non-small cell lung carcinoma (NSCLC) and human bronchial epithelial cells. Here, we investigate the reciprocal relationship between phosphorylation status of DNA-PKcs and EGFR-mediated radiation response. The data reveal that both the kinase activity of DNA-PKcs and radiation-induced phosphorylation of DNA-PKcs by the ataxia telangiectasia-mutated (ATM) kinase are critical prerequisites for EGFR-mediated radioresponse. Alanine substitutions at seven key serine/threonine residues in DNA-PKcs or inhibition of DNA-PKcs by NU7441 completely abrogated EGFR-mediated radioresponse and blocked EGFR binding. ATM deficiency or ATM inhibition with KU55933 produced a similar effect. Importantly, alanine substitution at an ATM-dependent DNA-PKcs phosphorylation site, T2609, was sufficient to block binding or radioresponse of EGFR. However, mutation of a DNA-PKcs autophosphorylation site, S2056 had no such effect indicating that DNA-PKcs autophosphorylation is not necessary for EGFR-mediated radioresponse. Our data reveal that in both NSCLCs and human bronchial epithelial cells, activating mutations in EGFR specifically abolished the DNA-PKcs phosphorylation at T2609, but not S2056. Our study underscores the critical importance of a reciprocal relationship between DNA-PKcs phosphorylation and EGFR-mediated radiation response and elucidates mechanisms underlying mutant EGFR-associated radiosensitivity in NSCLCs.


Subject(s)
DNA-Activated Protein Kinase/metabolism , ErbB Receptors/metabolism , Nuclear Proteins/metabolism , Phosphothreonine/metabolism , Radiation Tolerance , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Epithelial Cells/enzymology , Epithelial Cells/radiation effects , Humans , Mutant Proteins/metabolism , Mutation/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phosphorylation/radiation effects , Phosphoserine/metabolism , Protein Binding/radiation effects , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/radiation effects , Radiation, Ionizing , Structure-Activity Relationship , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
6.
Semin Radiat Oncol ; 20(3): 149-55, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20685577

ABSTRACT

The recently developed ability to interrogate genome-wide data arrays has provided invaluable insights into the molecular pathogenesis of lung cancer. These data have also provided information for developing targeted therapy in lung cancer patients based on the identification of cancer-specific vulnerabilities and set the stage for molecular biomarkers that provide information on clinical outcome and response to treatment. In addition, there are now large panels of lung cancer cell lines, both non-small-cell lung cancer and small-cell lung cancer, that have distinct chemotherapy and radiation response phenotypes. We anticipate that the integration of molecular data with therapy response data will allow for the generation of biomarker signatures that predict response to therapy. These signatures will need to be validated in clinical studies, at first retrospective analyses and then prospective clinical trials, to show that the use of these biomarkers can aid in predicting patient outcomes (eg, in the case of radiation therapy for local control and survival). This review highlights recent advances in molecular profiling of tumor responses to radiotherapy and identifies challenges and opportunities in developing molecular biomarker signatures for predicting radiation response for individual patients with lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Genomics/methods , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Biomarkers, Tumor , Humans , Treatment Outcome
7.
Carcinogenesis ; 31(10): 1889-96, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20663777

ABSTRACT

DNA double-strand breaks (DSBs) are the most deleterious lesion inflicted by ionizing radiation. Although DSBs are potentially carcinogenic, it is not clear whether complex DSBs that are refractory to repair are more potently tumorigenic compared with simple breaks that can be rapidly repaired, correctly or incorrectly, by mammalian cells. We previously demonstrated that complex DSBs induced by high-linear energy transfer (LET) Fe ions are repaired slowly and incompletely, whereas those induced by low-LET gamma rays are repaired efficiently by mammalian cells. To determine whether Fe-induced DSBs are more potently tumorigenic than gamma ray-induced breaks, we irradiated 'sensitized' murine astrocytes that were deficient in Ink4a and Arf tumor suppressors and injected the surviving cells subcutaneously into nude mice. Using this model system, we find that Fe ions are potently tumorigenic, generating tumors with significantly higher frequency and shorter latency compared with tumors generated by gamma rays. Tumor formation by Fe-irradiated cells is accompanied by rampant genomic instability and multiple genomic changes, the most interesting of which is loss of the p15/Ink4b tumor suppressor due to deletion of a chromosomal region harboring the CDKN2A and CDKN2B loci. The additional loss of p15/Ink4b in tumors derived from cells that are already deficient in p16/Ink4a bolsters the hypothesis that p15 plays an important role in tumor suppression, especially in the absence of p16. Indeed, we find that reexpression of p15 in tumor-derived cells significantly attenuates the tumorigenic potential of these cells, indicating that p15 loss may be a critical event in tumorigenesis triggered by complex DSBs.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/physiology , DNA Breaks, Double-Stranded , Neoplasms/etiology , Animals , Cells, Cultured , Chromosome Aberrations , Chromosome Deletion , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , DNA Repair , Genomic Instability , Humans , Mice
8.
Prostaglandins Other Lipid Mediat ; 92(1-4): 19-24, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20152925

ABSTRACT

2-Arachidonoylglycerol is oxygenated by cyclooxygenase-2 to form prostaglandin glyceryl esters. Previous work in this laboratory has suggested that PGE(2)-G activates a novel G protein-coupled receptor in a murine macrophage-like cell line, RAW 264.7. To probe the structural determinants for the putative receptor in RAW 264.7 cells, a panel of 10 analogs was tested for their ability to increase intracellular calcium. These analogs included PGE(2)- and PGF(2alpha)-ethanolamide, 4 PGE(2) glyceryl ester analogs, and 4 PGF(2alpha) glyceryl ester analogs. The glyceryl ester analogs differed in the positioning of the hydroxyl groups in the glycerol moiety and the type of linker (ester, amide, or thioester) of the prostaglandin to the glycerol moiety. Compounds were also evaluated in a human non-small cell lung cancer cell line (H1819). The glycerol moiety was required for the calcium response. All glyceryl ester analogs but not ethanolamides caused a concentration-dependent increase in calcium levels in both RAW 264.7 and H1819 cells. An amide or ester linkage was preferable to a thioester linkage. The EC(50) values did not significantly change when the positioning of the hydroxyls was varied. This calcium response induced by the glyceryl ester analogs appears to be independent of the putative hydrolysis products, PGE(2) and PGF(2alpha), and appears to represent a novel signaling pathway.


Subject(s)
Calcium Signaling/drug effects , Dinoprost/chemistry , Dinoprost/pharmacology , Dinoprostone/chemistry , Dinoprostone/pharmacology , Esters/chemistry , Animals , Cell Line, Tumor , Ethanolamine/chemistry , Humans , Mice , Structure-Activity Relationship
9.
Clin Cancer Res ; 13(22 Pt 1): 6555-60, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-18006754

ABSTRACT

The epidermal growth factor receptor (EGFR), which is frequently expressed in tumors of epithelial origin, is an important determinant of tumor responses to ionizing radiation. Elevated EGFR expression and activity frequently correlate with tumor resistance to radiotherapy in patients. EGFR is thought to confer tumor resistance to radiation through the activation of survival and cell proliferation pathways. Recent discoveries have identified a novel radioprotective function of EGFR which involves the radiation-induced nuclear translocation of the receptor and its interactions with the DNA-dependent protein kinase, a key component of the nonhomologous end-joining DNA repair pathway. Targeting the DNA repair function of EGFR may serve as a therapeutic model for sensitizing tumors to radiotherapy in patients.


Subject(s)
DNA Repair , DNA-Activated Protein Kinase/metabolism , ErbB Receptors/metabolism , Neoplasms/radiotherapy , Radiation Tolerance , Animals , DNA/radiation effects , ErbB Receptors/genetics , Humans , Mice , Neoplasms/enzymology , Radiation Tolerance/genetics
10.
Cancer Res ; 67(11): 5267-74, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545606

ABSTRACT

The epidermal growth factor receptor (EGFR) is an important determinant of radioresponse, whose elevated expression and activity frequently correlates with radioresistance in several cancers, including non-small cell lung carcinoma (NSCLC). We reported recently that NSCLC cell lines harboring somatic, activating mutations in the tyrosine kinase domain (TKD) of the EGFR exhibit significant delays in the repair of DNA double-strand breaks (DSB) and poor clonogenic survival in response to radiation. Here, we explore the mechanisms underlying mutant EGFR-associated radiosensitivity. In three representative NSCLC cell lines, we show that, unlike wild-type (WT) EGFR, receptors with common oncogenic TKD mutations, L858R or DeltaE746-E750, are defective in radiation-induced translocation to the nucleus and fail to bind the catalytic and regulatory subunits of the DNA-dependent protein kinase (DNA-PK), a key enzyme in the nonhomologous end-joining repair pathway. Moreover, despite the presence of WT EGFR, stable exogenous expression of either the L858R or the DeltaE746-E750 mutant forms of EGFR in human bronchial epithelial cells significantly delays repair of ionizing radiation (IR)-induced DSBs, blocks the resolution of frank or microhomologous DNA ends, and abrogates IR-induced nuclear EGFR translocation or binding to DNA-PK catalytic subunit. Our study has identified a subset of naturally occurring EGFR mutations that lack a critical radioprotective function of EGFR, providing valuable insights on how the EGFR mediates cell survival in response to radiation in NSCLC cell lines.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/radiotherapy , ErbB Receptors/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/radiotherapy , Mutation , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Nucleus/enzymology , DNA Damage , DNA Repair , DNA, Neoplasm/genetics , DNA, Neoplasm/radiation effects , DNA-Activated Protein Kinase/metabolism , ErbB Receptors/metabolism , Humans , Lung Neoplasms/genetics , Phosphorylation , Protein Structure, Tertiary , Radiation Tolerance/genetics , Transfection
11.
Cancer Res ; 66(19): 9601-8, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17018617

ABSTRACT

Non-small cell lung cancers (NSCLCs) bearing mutations in the tyrosine kinase domain (TKD) of the epidermal growth factor receptor (EGFR) often exhibit dramatic sensitivity to the EGFR tyrosine kinase inhibitors gefitinib and erlotinib. Ionizing radiation (IR) is frequently used in the treatment of NSCLC, but little is known how lung tumor-acquired EGFR mutations affect responses to IR. Because this is of great clinical importance, we investigated and found that clonogenic survival of mutant EGFR NSCLCs in response to IR was reduced 500- to 1,000-fold compared with wild-type (WT) EGFR NSCLCs. Exogenous expression of either the L858R point mutant or the DeltaE746-E750 deletion mutant form of EGFR in immortalized human bronchial epithelial cells, p53 WT NSCLC (A549), or p53-null NSCLC (NCI-H1299) resulted in dramatically increased sensitivity to IR. We show that the majority of mutant EGFR NSCLCs, including those that contain the secondary gefitinib resistance T790M mutation, exhibit characteristics consistent with a radiosensitive phenotype, which include delayed DNA repair kinetics, defective IR-induced arrest in DNA synthesis or mitosis, and pronounced increases in apoptosis or micronuclei. Thus, understanding how activating mutations in the TKD domain of EGFR contribute to radiosensitivity should provide new insight into effective treatment of NSCLC with radiotherapy and perhaps avoid emergence of single agent drug resistance.


Subject(s)
Beta Particles , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/physiology , Gamma Rays , Genes, erbB-1 , Lung Neoplasms/genetics , Neoplasm Proteins/physiology , Apoptosis/genetics , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , DNA Repair/genetics , DNA Replication/genetics , DNA Replication/radiation effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/chemistry , Exons/genetics , Gefitinib , Genes, p53 , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mutation , Neoplasm Proteins/chemistry , Protein Structure, Tertiary , Quinazolines/pharmacology , Radiation Tolerance/genetics , Recombinant Fusion Proteins/physiology , Sequence Deletion , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay
12.
Proc Natl Acad Sci U S A ; 101(7): 1840-5, 2004 Feb 17.
Article in English | MEDLINE | ID: mdl-14766978

ABSTRACT

Glyceryl prostaglandins (PG-Gs) are generated by the oxygenation of the endocannabinoid, 2-arachidonylglycerol, by cyclooxygenase 2. The biological consequences of this selective oxygenation are uncertain because the cellular activities of PG-Gs have yet to be defined. We report that the glyceryl ester of PGE(2), PGE(2)-G, triggers rapid, concentration-dependent Ca(2+) accumulation in a murine macrophage-like cell line, RAW264.7. Ca(2+) mobilization is not observed after addition of PGE(2), PGD(2)-G, or PGF(2alpha)-G but is observed after addition of PGF(2alpha). Moreover, PGE(2)-G, but not PGE(2), stimulates a rapid but transient increase in the levels of inositol 1,4,5-trisphosphate (IP(3)) as well as the membrane association and activation of PKC. PGE(2)-G induces a concentration-dependent increase in the levels of phosphorylated extracellular signal regulated kinases 1 and 2 through a pathway that requires the activities of PKC, IP(3) receptor, and phospholipase C beta. The results indicate that PGE(2)-G triggers Ca(2+) mobilization, IP(3) synthesis, and activation of PKC in RAW264.7 macrophage cells at low concentrations. These responses are independent of the hydrolysis of PGE(2)-G to PGE(2).


Subject(s)
Calcium/metabolism , Dinoprostone/chemistry , Dinoprostone/pharmacology , Signal Transduction/drug effects , Animals , Calcium Signaling/drug effects , Cell Line , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Genes, Reporter , Inositol 1,4,5-Trisphosphate/metabolism , Macrophages , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Receptors, Prostaglandin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...