Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters










Publication year range
1.
Curr Cancer Drug Targets ; 12(2): 97-106, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22165966

ABSTRACT

Non-small cell lung carcinoma (NSCLC) is the leading type of lung cancer; smoking is a documented risk factor. Nicotinic acetylcholine receptor (nAChR)-mediated intracellular signaling in response to nicotine has recently been implicated in the growth regulation of NSCLC. In the current study nude mice carrying xenografts of the human lung NSCLC cell lines NCI-H322 or NCI-H441 were used as animal models. Nicotine administration and gamma aminobutyric acid (GABA) treatment lasted for 30 days. Catecholamines, cortisol, GABA, and cAMP were analyzed in blood and tumor tissues by immunoassays. Expression of nicotinic receptors and effector proteins in the xenografts was assessed by Western blotting. Our data indicate that nicotine stimulated the growth of NSCLC xenografts via modulation of nAChR upregulation and activation of cAMP signaling. The nicotine-treated group showed an enhanced level of stress neurotransmitters and second messenger cAMP in serum, blood cellular fraction, and xenograft tissues. Activation of critical proteins in the oncogenic pathway, including CREB, ERK, Akt, and Src, and upregulation of α-4 and α-7 subunits of nAChR provided mechanistic insight for the observed stimulatory effect of nicotine. Interestingly, GABA, being an antagonist to cAMP signaling, showed a promising intervention by reversing the stimulatory effect of nicotine on cancer growth and all signaling pathways. GABA has potential to lower the risk of NSCLC among smokers and could be used to enhance the clinical outcome of standard cancer intervention strategies.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Nicotine/antagonists & inhibitors , Xenograft Model Antitumor Assays , gamma-Aminobutyric Acid/pharmacology , Animals , Cell Division/drug effects , Humans , Male , Mice , Mice, Nude , Nicotine/pharmacology , Receptors, Nicotinic/drug effects
2.
Int J Cancer ; 119(7): 1547-52, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16671086

ABSTRACT

We have shown that the tobacco nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is an agonist for -adrenergic receptors (beta-ARs) and increased DNA synthesis of human lung adenocarcinoma cells with features of bronchiolar Clara cells by binding to these receptors. Using a cell line derived from a human pulmonary adenocarcinoma with Clara cell phenotype (PACC) and immortalized human small airway epithelial cells (HPLD1), the putative cells of origin of this cancer type, our current studies have analyzed signaling initiated by binding of NNK to the beta 1-AR. NNK upregulated ERK1/2 and CREB/ATF-1 phosphorylation in a PKA-dependent manner in both cell lines. This response was further increased by transient overexpression of the beta 1-AR. Pre-exposure of cells to the selective beta 1-AR antagonist, atenolol, attenuated the stimulatory effects of NNK, suggesting the latter upregulated ERK1/2 and CREB/ATF-1 via this receptor. In vivo labeling and immunoprecipitation assays revealed that NNK phosphorylated the epidermal growth factor receptor (EGFR) at tyrosine residues, 991, 1068 and 1173, an effect inhibited by atenolol. The inhibitor of EGFR-specific tyrosine kinases, AG1478, reduced NNK ability to stimulate ERK1/2 and CREB/ATF-1. Genomic analysis of the exons 18-21 of the EGFR genes showed that no mutations were present in either gene. Collectively, our data provide evidence, for the first time, that NNK targets ERK1/2 and CREB/ATF-1 proteins via dual signaling involving beta 1-AR and EGFR pathways in PACCs and their putative cells of origin.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/pathology , Nitrosamines/pharmacology , Nuclear Proteins/metabolism , Receptors, Adrenergic, beta-1/metabolism , Activating Transcription Factor 1 , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Cell Line , DNA/biosynthesis , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , ErbB Receptors/genetics , Humans , Lung/cytology , Lung Neoplasms/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation/genetics , Phosphorylation/drug effects , Regulatory Factor X Transcription Factors , Signal Transduction/drug effects , Transcription Factors
3.
Eur J Cancer ; 41(17): 2745-53, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16239108

ABSTRACT

Smoking is a risk factor for lung cancer, chronic obstructive pulmonary disease, chronic bronchitis and asthma. The chronic lung diseases are also a predisposing factor for the development of lung cancer. Glucocorticoids are used for the management of chronic lung diseases because of their anti-inflammatory activity. These drugs also have anti-tumourigenic effects in mouse models of lung cancer. Glucocorticoids are frequently used as co-treatment with cancer therapy. Using the human pulmonary adenocarcinoma (PAC) cell line NCI-H322 with features of bronchiolar Clara cells, and immortalised human small airway epithelial cells, our data show that the glucocorticoid dexamethasone increased cell proliferation in MTT assays in a PKA-dependent manner. Dexamethasone significantly increased intracellular cAMP in direct immunoassays. Immunoblot analysis revealed increased phosphorylation of ERK1/2 and of the transcription factor CREB in response to dexamethasone. These data suggest that glucocorticoids could have tumour promoting activity on a sub-set of human PAC.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Bronchi/pathology , Cyclic AMP-Dependent Protein Kinases/pharmacology , Dexamethasone/pharmacology , Lung Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Humans , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology
4.
J Cancer Res Clin Oncol ; 128(10): 525-32, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12384795

ABSTRACT

PURPOSE: Pancreatic cancer is the fourth leading cause of cancer death in men and women. Smoking is a documented risk factor for pancreatic cancer, and the risk is increased in smokers who also consume alcohol. Arachidonic acid (AA)-metabolizing enzymes have been implicated in aggressive clinical behavior of pancreatic cancer while mutations in the Ki- ras gene have been associated with prolonged survival and responsiveness to therapy. Using a hamster model of exocrine pancreatic cancer induced by transplacental exposure to ethanol and the tobacco-carcinogen NNK, we have analyzed these tumors for mutations in the ras and p53 genes and tested the modulating effects of the COX inhibitor, ibuprofen, and the FLAP inhibitor, MK886, on the development of pancreatic cancer in this animal model. METHODS: Hamsters were given 10% ethanol in the drinking water from the fifth to the last day of their pregnancy and a single dose of NNK on the last day. Starting at 4 weeks of age, groups of offspring were given either the COX inhibitor ibuprofen (infant Motrin oral suspension) or the FLAP-inhibitor MK886 (dissolved in carboxymethylcellulose orally) for life while a group of offspring not receiving any treatment served as positive controls. RESULTS: None of the induced pancreatic cancers demonstrated mutations in the Ki-, N-, or H- ras or p53 genes. The development of pancreatic cancer in offspring who had been given ibuprofen or MK886 was reduced by 50% or 30%, respectively. CONCLUSION: In conjunction with the documented over-expression of COX-2 and LOX in human pancreatic cancer, our findings suggest an important role of the AA-cascade in the genesis of this cancer type and indicate that pharmacological or dietary measures that reduce AA-metabolism may be useful for the prevention and clinical management of pancreatic cancer.


Subject(s)
Adenocarcinoma/prevention & control , Cyclooxygenase Inhibitors/therapeutic use , Ibuprofen/therapeutic use , Indoles/therapeutic use , Lipoxygenase Inhibitors/therapeutic use , Pancreatic Neoplasms/prevention & control , Adenocarcinoma/chemically induced , Adenocarcinoma/genetics , Animals , Arachidonic Acid/metabolism , Cricetinae , DNA Primers/chemistry , Ethanol/toxicity , Female , Genes, p53/genetics , Genes, ras/genetics , Male , Maternal-Fetal Exchange/drug effects , Mesocricetus , Nitrosamines/toxicity , Pancreas/drug effects , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Pregnancy
5.
Int J Oncol ; 21(1): 153-7, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12063562

ABSTRACT

Adenocarcinoma of the mammary gland is the leading type of cancer in women. Among these breast cancers those that are estrogen-responsive respond well to existing therapeutic regimens while estrogen non-responsive cancers metastasize widely, demonstrate a high relapse rate, and respond poorly to therapy. Over-expression of the arachidonic acid-metabolizing enzymes cyclooxygenase-2 and lypoxygenases is frequently observed in breast cancer, particularly the non-estrogen-responsive type, suggesting a role of the arachidonic acid (AA) cascade in the growth regulation of these malignancies. Adenocarcinomas of the lungs, pancreas and colon also frequently over-express AA-metabolizing enzymes, and recent evidence suggests that the growth-regulating AA-cascade in these malignancies is under beta-adrenergic control. Our current experiments have therefore tested the hypothesis that in analogy to these findings adenocarcinomas of the breast are also regulated by beta-adrenergic receptors via stimulation of the AA-cascade. Analysis of DNA synthesis by [3H]-thymidine incorporation assays in three estrogen-responsive and three estrogen non-responsive cell lines derived from human breast cancers demonstrated a significant reduction in DNA synthesis by beta-blockers and inhibitors of cyclooxygenase or lipoxygenases in all cell lines. Analysis of AA-release in one of the most responsive cell lines demonstrated a time-dependent increase in AA-release in response to the beta-adrenergic agonist isoproterenol. Analysis by RT-PCR revealed expression of beta2-adrenergic receptors in all cell lines whereas beta1-adrenergic receptors were not found in two of the estrogen non-responsive cell lines. Our data suggest that a significant subset of human breast cancers is under control of beta-adrenergic receptors via stimulation of the AA-cascade. These findings open up novel avenues for the prevention and clinical management of breast cancer, particularly the non-estrogen-responsive types. Moreover, our findings suggest that cardiovascular disease and adenocarcinomas in a variety of organ systems, including the breast may share common risk factors and benefit from similar preventive and treatment strategies.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Arachidonic Acid/metabolism , Breast Neoplasms/pathology , Cyclooxygenase Inhibitors/pharmacology , Lipoxygenase Inhibitors/pharmacology , Adrenergic beta-Agonists/pharmacology , Arachidonate 5-Lipoxygenase/metabolism , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , DNA/biosynthesis , DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Estrogens/pharmacology , Female , Humans , Isoenzymes/antagonists & inhibitors , Isoproterenol/pharmacology , Membrane Proteins , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Prostaglandin-Endoperoxide Synthases , RNA, Messenger/metabolism , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
6.
Am J Physiol Heart Circ Physiol ; 281(5): H1946-54, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11668055

ABSTRACT

Smoking causes endothelial cell (EC) injury; however, neither the components of cigarette smoke nor the mechanisms responsible for this injury are understood. The nitrosated derivative of nicotine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), has been implicated in the carcinogenic effects of tobacco; however, the effects of NNK on the cardiovascular system are largely unknown. NNK binds to beta1- and beta2-adrenergic receptors. Because beta-adrenergic receptor activation causes arachidonic acid (AA) release and cellular injury, we postulated that NNK causes EC injury by a mechanism that involves beta-adrenergic-mediated release of AA. NNK stimulated [3H]AA release from ECs, and this effect was mediated by both beta1- and beta2-adrenergic receptors because pretreatment with atenolol or ICI 118,551 inhibited the response. NNK also induced EC apoptosis, as measured by terminal deoxyribonucleotide transferase-mediated dUTP nick-end labeling and annexin V staining. NNK-mediated apoptosis was attenuated by pretreatment with atenolol or ICI 118,551. Furthermore, depletion of cellular AA by incubation with eicosapentaenoic acid abolished the apoptotic effect of NNK. These data suggest that NNK causes EC apoptosis by a mechanism that involves beta1- and beta2-adrenergic receptor-mediated release of AA.


Subject(s)
Apoptosis/drug effects , Carcinogens/pharmacology , Endothelium, Vascular/cytology , Nitrosamines/pharmacology , Animals , Aorta/cytology , Arachidonic Acid/pharmacokinetics , Arteriosclerosis/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , In Situ Nick-End Labeling , Nicotine/pharmacokinetics , Receptors, Adrenergic, beta/metabolism , Smoking/adverse effects , Swine , Tritium
7.
Int J Oncol ; 19(3): 445-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11494020

ABSTRACT

Pulmonary adenocarcinoma (PAC) is the leading type of lung cancer and is highly resistant to conventional cancer therapy. A better understanding of the regulatory mechanisms which control the growth of this deadly malignancy are urgently needed to develop more effective cancer intervention strategies. Recent studies have shown that PAC frequently overexpresses cyclooxygenase-2 (COX-2). This enzyme converts arachidonic acid (AA) into several metabolites, some of which have been identified as modulators of mitogenesis and apoptosis. Accordingly, the AA cascade and COX-2 are currently widely studied as potential targets for lung cancer prevention. Recent studies by our research group have shown that cell lines derived from human PACs express beta1- and beta2-adrenergic receptors, which regulate the release of AA and DNA synthesis. Moreover, we have demonstrated that an antagonist for beta-adrenergic receptors or aspirin inhibited the development of experimentally induced PAC in a hamster model. These findings suggest that beta-adrenergic receptors may serve as upstream regulators of AA and COX-2-mediated PAC growth. However, no information is currently available on the expression of beta-adrenergic receptors and its possible correlation with the expression of COX-2 in tissue samples from human PAC, casting some doubt on the significance of these findings in vitro and in an animal model. In the current study, we have therefore analyzed tissue samples of human PACs for the expression of beta1-and beta2-adrenergic receptors as well as COX-2 by reverse transcription polymerase chain reaction (RT-PCR) or immunohistochemistry. Our data show that seven out of eight samples co-expressed COX-2 and one or both of these beta-adrenergic receptors, supporting the experimental evidence for a functional link between these neurotransmitter receptors and the AA cascade in the regulation of human PAC.


Subject(s)
Adenocarcinoma/metabolism , Isoenzymes/metabolism , Lung Neoplasms/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Cyclooxygenase 2 , DNA Primers/chemistry , Epithelial Cells/pathology , Humans , Immunoenzyme Techniques , Isoenzymes/genetics , Membrane Proteins , Paraffin Embedding , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-2/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Carcinogenesis ; 22(3): 473-9, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238189

ABSTRACT

Exocrine ductal carcinoma of the pancreas has been associated with smoking, and the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) causes this cancer type in laboratory rodents. Current knowledge on the growth regulation of this malignancy is extremely limited. Recent studies have shown overexpression of cyclooxygenase 2 (COX 2) and 5-lipoxygenase (5-lipox) in exocrine pancreatic carcinomas, suggesting a potential role of the arachidonic acid (AA) cascade in the regulation of this cancer type. In support of this interpretation, our data show high basal levels of AA release in two human cell lines derived from exocrine ductal pancreatic carcinomas. Both cell lines expressed m-RNA for beta2-adrenergic receptors and beta1-adrenergic receptors. Radio-receptor assays showed that beta2-adrenergic receptors predominated over beta1-adrenergic receptors. beta2-Adrenergic antagonist ICI118,551 significantly reduced basal AA release and DNA synthesis when the cells were maintained in complete medium. DNA synthesis of the cell line (Panc-1) with an activating point mutation in codon 12 of the ki-ras gene was significantly stimulated by NNK when cells were maintained in complete medium and this response was inhibited by the beta-blocker ICI118,551, the COX-inhibitor aspirin, or the 5-lipox-inhibitor MK-886. The cell line without ras mutations (BXPC-3) did not show a significant response to NNK in complete medium. When the assays were conducted in serum-free medium, both cell lines demonstrated increased DNA synthesis in response to NNK, an effect inhibited by the beta2-blocker, aspirin, or MK-886. Panc-1 cells were more sensitive to the stimulating effects of NNK and less responsive to the inhibitors than BXPC-3 cells. Our findings are in accord with a recent report which has identified NNK as a beta-adrenergic agonist and suggest beta-adrenergic, AA-dependent regulatory pathways in pancreatic cancer as a novel target for cancer intervention strategies.


Subject(s)
Adenocarcinoma/pathology , Pancreatic Neoplasms/pathology , Receptors, Adrenergic, beta/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adrenergic beta-Antagonists/pharmacology , Arachidonic Acid/metabolism , Aspirin/pharmacology , Base Sequence , DNA Primers , Enzyme Inhibitors/pharmacology , Genes, ras , Humans , Indoles/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Propanolamines/pharmacology , Radioligand Assay , Receptors, Adrenergic, beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
9.
J Cancer Res Clin Oncol ; 127(12): 707-17, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11768610

ABSTRACT

OBJECTIVE: Small cell lung carcinoma (SCLC) expresses phenotypic features of pulmonary neuroendocrine cells and demonstrates a strong etiologic association with smoking. SCLC cell lines express a Raf-1-dependent mitogenic signal transduction pathway, which is thought to transduce the mitogenic signals initiated by neuropeptide autocrine growth factors. Recent studies have identified the tobacco-specific carcinogenic nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) as a site-selective high-affinity agonist for the alpha7 nicotinic acetylcholine receptor (alpha7 nAChR), which regulates the growth of a significant subset of SCLC in vitro by stimulating the release of the autocrine growth factor serotonin. The purpose of this study was to identify signaling events initiated by binding of NNK to the alpha7 nAChR. DESIGN: We have used a human SCLC cell line and fetal hamster pulmonary neuroendocrine cells with in vitro kinase activation assays and western blots to assess the levels of expression and activation of Raf-1, MAPK and c-myc to address this issue. RESULTS: Our data show that NNK activates the Raf-1, MAP kinase pathway, resulting in phosphorylation of c-myc. The activation of this signal transduction pathway by NNK was inhibited by the site-selective antagonist for the alpha7 nAChR alpha-bungarotoxin (alpha-BTX) or by the serotonin reuptake inhibitor imipramine, suggesting that the responses to NNK were mediated by nicotinic receptor-initiated release of serotonin. Accordingly, NNK-induced 5-HT release was blocked by alpha-BTX while NNK-induced DNA synthesis was inhibited by alpha-BTX, imipramine, the PKC inhibitor sphingosine or the MEK inhibitor PD98059. SCLC cells demonstrated high basal levels of 5-HT release, DNA synthesis, and over-expressed Raf-1 and MAPK protein suggesting the constitutive activation of an upstream regulator such as the alpha7 nAChR. CONCLUSION: Our findings link, for the first time, the stimulation of a nicotinic acetylcholine receptor by a cancer-causing agent with the activation of a Raf-1/MAPK/c-myc signaling pathway. Furthermore, our data suggest that serotonin uptake inhibitors may protect against the development or be useful in the clinical management of SCLC.


Subject(s)
Carcinogens/toxicity , Carcinoma, Small Cell/chemically induced , Lung Neoplasms/chemically induced , Lung/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurosecretory Systems/drug effects , Nitrosamines/toxicity , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-raf/physiology , Receptors, Nicotinic/physiology , Animals , Bungarotoxins/pharmacology , Carcinoma, Small Cell/metabolism , Cricetinae , Enzyme Activation/drug effects , Humans , Imipramine/pharmacology , Lung/metabolism , Lung Neoplasms/metabolism , Neurosecretory Systems/metabolism , Phosphorylation , Polymerase Chain Reaction , Serotonin/physiology , Tumor Cells, Cultured
10.
J Cancer Res Clin Oncol ; 126(11): 624-30, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11079726

ABSTRACT

OBJECTIVE: Lung cancer is the leading cause of cancer death in industrialized countries. Pulmonary adenocarcinoma (PAC) is the most common histologic type of lung cancer, and it is reproducibly induced by the tobacco-specific nitrosamine 4-(methylnitrosamino)- 1-(3-pyridyl)-1-butanone (NNK) in laboratory rodents. We have recently shown that the growth of cell lines derived from human PACs is controlled by beta-adrenergic receptors, and that NNK is a high affinity agonist for this receptor family. DESIGN: In the current study, we have tested the relevance of these in in vitro findings for in vivo NNK-induced lung tumorigenesis, using a well-established hamster model of NNK-induced PAC. RESULTS: Our experiments demonstrate a significant increase in NNK-induced PAC multiplicity in animals chronically exposed to the beta-adrenergic agonist epinephrine or theophylline which causes intracellular accumulation of the beta-adrenergic second messenger cAMP. On the other hand, our data show that administration of the beta-adrenergic antagonist propranolol prior to each NNK injection significantly inhibited the development of PACs. DISCUSSION: Our findings support the hypothesis that the development of tobacco-associated PAC may be modulated by beta-adrenergic agents, and that the interaction of NNK with beta-adrenergic receptors contributes to the genesis of this histologic lung cancer type.


Subject(s)
Adenocarcinoma/chemically induced , Adenocarcinoma/prevention & control , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Lung Neoplasms/chemically induced , Lung Neoplasms/prevention & control , Nitrosamines/pharmacology , Adrenergic beta-Agonists/toxicity , Animals , Carcinogens/pharmacology , Carcinogens/toxicity , Cricetinae , Drug Synergism , Epinephrine/pharmacology , Epinephrine/toxicity , Male , Mesocricetus , Nitrosamines/toxicity , Propranolol/pharmacology , Receptors, Adrenergic, beta/physiology , Theophylline/pharmacology , Theophylline/toxicity
11.
Toxicol Sci ; 56(2): 313-23, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10910989

ABSTRACT

Previous studies from this laboratory have demonstrated that administration of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) to pregnant hamsters results in tumors in the offspring. Whereas treatment with NNK alone caused mainly tumors in the respiratory tract of the treated offspring, cotreatment with ethanol (EtOH) and NNK shifted the site of tumor formation to the pancreas. In order to determine potential mechanisms for the cocarcinogenic effects of EtOH, the levels of NNK metabolites and expression of various CYPs implicated in the metabolic activation of NNK were determined in fetal liver and pancreas. NNK and its metabolite, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), were detected at low and variable levels in the fetal liver and pancreas, with an NNAL to NNK ratio greater than 20 in both organs. EtOH had no effect on the amount of metabolites found in either organ. Results obtained with the fetal liver samples, which served as a positive control, correlated very well with our previous studies demonstrating low levels of expression of several CYP isozymes at both the protein and RNA level. Western blot analysis showed low but detectable levels of CYP1A1, barely detectable levels of CYP2E1, and an absence of CYP1A2 and 2B family members in the fetal pancreas. RNA transcripts were undetectable by ribonuclease protection in the fetal pancreas, although readily seen in fetal liver samples. Treatment with NNK, EtOH, or both NNK and EtOH had small and variable effects on the levels of metabolism of NNK and expression of the isozymes. These findings suggest that alternative mechanisms may be responsible for transplacentally induced tumors in this model system.


Subject(s)
Carcinogens/toxicity , Cytochrome P-450 Enzyme System/biosynthesis , Ethanol/toxicity , Fetal Diseases/enzymology , Nitrosamines/toxicity , Pancreatic Neoplasms/enzymology , Animals , Blotting, Western , Cricetinae , DNA Adducts/analysis , Female , Fetal Diseases/chemically induced , Liver/metabolism , Mesocricetus , Nitrosamines/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/chemically induced , Pregnancy
12.
Eur J Pharmacol ; 393(1-3): 265-77, 2000 Mar 30.
Article in English | MEDLINE | ID: mdl-10771023

ABSTRACT

Pulmonary neuroendocrine cells function as hypoxia-sensitive chemoreceptors, and they release peptides and biogenic amines that are important mediators of pulmonary neonatal adaptation. Some of these products additionally act as autocrine growth factors. Increased numbers of pulmonary neuroendocrine cells have been observed in several smoking-associated pediatric lung disorders such as bronchopulmonary dysplasia, cystic fibrosis, sudden infant death syndrome, and asthma. Disturbed pulmonary neuroendocrine function has been implicated in the etiology of this disease complex. One of the most common smoking-associated lung cancer types, small cell lung carcinoma, expresses phenotypic and functional features of pulmonary neuroendocrine cells. We, as well as others, have shown that the release of the autocrine growth factors 5-hydroxytryptamine (5-HT, serotonin) and mammalian bombesin/gastrin releasing peptide (MB/GRP) by cell lines derived from human small cell lung carcinoma or fetal hamster pulmonary neuroendocrine cells are regulated by a neuronal nicotinic acetylcholine receptor comprised of alpha(7) subunits. In radio-receptor assays, nicotine and the nicotine-derived carcinogenic nitrosamines NNNN. Binding of nicotine or NNK to the alpha(7) receptor resulted in calcium influx and overexpression and activation of the serine-threonine protein kinase Raf-1. In turn, this event lead to overexpression and activation of the mitogen activated (MAP) kinases extracellular signal regulated kinase 1 (ERK1) and extracellular signal regulated kinase 2 (ERK2) and stimulation of DNA synthesis accompanied by an increase in cell numbers in fetal pulmonary neuroendocrine cells and small cell carcinoma cells. Exposure of fetal pulmonary neuroendocrine cells for 6 days to NNK caused a prominant up-regulation of Raf-1. Our findings suggest that chronic exposure to nicotine and NNK in pregnant women who smoke may up-regulate the alpha(7) nicotinic receptor as well as components of its associated mitogenic signal transduction pathway, thus increasing the susceptibilities of the infants for the development of pediatric lung disorders. Similarly, up-regulation of one or several components of this nicotinic receptor pathway in smokers may be an important factor for the development of small cell lung carcinoma.


Subject(s)
Lung Neoplasms/chemically induced , MAP Kinase Signaling System , Nicotiana/adverse effects , Nitrosamines/toxicity , Plants, Toxic , Receptors, Nicotinic/metabolism , Animals , Binding, Competitive , Cell Line , Child , Child, Preschool , Cricetinae , Female , Humans , Lung Diseases/chemically induced , Lung Diseases/metabolism , Lung Neoplasms/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Nicotinic/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism , Tumor Cells, Cultured , alpha7 Nicotinic Acetylcholine Receptor
13.
Exp Lung Res ; 26(2): 121-35, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10742926

ABSTRACT

Pulmonary neuroendocrine cells (PNEC) produce neuropeptides and biogenic amines which regulate pulmonary vasoconstriction and bronchoconstriction. Increased numbers of PNEC along with elevated levels of their products have been consistently observed in a variety of pediatric pulmonary diseases, some of which are etiologically linked with prenatal exposure to cigarette smoke, and all of which are exacerbated by passive smoking. The objective of our studies was to characterize the autonomic regulation of these cells and to explore a potential direct interaction of tobacco-specific toxicants with these regulatory pathways. Using reverse transcription polymerase chain reaction (RT-PCR), radioreceptor assays, flow cytometry, enzyme-linked immunosorbent assay (ELISA), and cell proliferation assays, we found that the release of serotonin from fetal PNEC is regulated by a neuronal alpha 7-nicotinic acetylcholine receptor (alpha 7-nAChR) via a Ca(2+)-dependent mechanism. The tobacco-specific toxicants nicotine and 4-(methylnitrosamino)-1-(-3-pyridine)-1-butanone (NNK) bound with high affinity to this receptor, resulting in influx of Ca2+, release of serotonin, and stimulation of DNA synthesis. Chronic stimulation of the alpha 7-nAChR in the fetal lungs by prenatal exposure to cigarette smoke may contribute to the development of smoking-related pediatric pulmonary disease, whereas postnatal chronic exposure to environmental smoke may exacerbate these pediatric diseases via direct interaction with this receptor.


Subject(s)
Fetus/drug effects , Lung/drug effects , Neurosecretory Systems/drug effects , Nicotiana/toxicity , Plants, Toxic , Receptors, Nicotinic/drug effects , Animals , Cell Division , Cells, Cultured , Cricetinae , DNA/biosynthesis , Fetus/pathology , Fetus/physiopathology , Humans , Infant , Lung/pathology , Lung/physiopathology , Lung Diseases/etiology , Neurosecretory Systems/pathology , Neurosecretory Systems/physiopathology , Receptors, Nicotinic/physiology , Serotonin/metabolism , Smoking/adverse effects
14.
Int J Oncol ; 16(3): 513-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10675483

ABSTRACT

The nicotine-derived tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) induces lung cancer in all animal species tested and is thought to contribute significantly to the high lung cancer burden associated with smoking. NNK has recently been identified as a high affinity ligand for neuronal nicotinic acetylcholine receptors comprised of alpha7 subunits (alpha7 nAChR), and expressed in human small cell lung carcinoma (SCLC). As agonist-binding to this receptor in mammalian cells often results in membrane depolarization and activation of voltage-operated Ca2+-channels (VOCCs), we hypothesized that NNK may exert similar effects in SCLC. Using flow cytometry to monitor the influx of Ca2+, reverse transcription polymerase chain reaction (RT-PCR) to determine the expression of VOCC-specific messenger RNA, as well as analysis of DNA synthesis or determination of cell number, our data demonstrate that binding of NNK to the alpha7 nAChR in SCLC cells caused influx of Ca2+ via VOCCs of the L-, N-, and P-type. In turn, this led to a significant increase in DNA synthesis and cell number which was inhibited by a site-selective antagonist for the alpha7 nAChR and by Ca2+-channel blockers of the L-, N-, or P-types of VOCCs. Our findings suggest that the chronic activation of VOCC-mediated Ca2+ influx by NNK in smokers is an important event that may affect numerous Ca2+-dependent signal transduction pathways, thus contributing significantly to the development of SCLC.


Subject(s)
Calcium Channels/metabolism , Carcinogens/toxicity , Carcinoma, Small Cell/metabolism , Lung Neoplasms/metabolism , Nitrosamines/toxicity , Humans , Nicotine/toxicity , Tumor Cells, Cultured
15.
Cancer Res ; 59(18): 4510-5, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10493497

ABSTRACT

Lung cancer is the leading cause of death in the United States, and it demonstrates a strong etiological association with smoking. The nicotine-derived nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) reproducibly induces pulmonary adenocarcinomas (ACs) in laboratory rodents and is considered an important contributing factor to the high lung cancer burden observed in smokers. It has been shown that the development of NNK-induced ACs in mice is reduced by inhibitors of cyclooxygenase and lipoxygenase and that the growth of human AC cell lines is regulated by beta-adrenergic receptors. On the basis of structural similarities of NNK with classic beta-adrenergic agonists, we tested the hypothesis that NNK stimulates the growth of human AC cells via agonist-binding to beta-adrenergic receptors, resulting in the release of arachidonic acid (AA). In support of this hypothesis, radioreceptor assays with transfected CHO cell lines stably expressing the human beta1- or beta2-adrenergic receptor demonstrated high affinity binding of NNK to each of these receptors. Two human AC cell lines expressed beta1- and beta2-adrenergic receptors by reverse transcription-PCR and responded to NNK with the release of AA and an increase in DNA synthesis. Beta-adrenergic antagonists completely blocked the release of AA and increase in DNA synthesis. The cyclooxygenase inhibitor aspirin and the 5-lipoxygenase inhibitor MK-886 both partially inhibited DNA synthesis in response to NNK. Our findings identify the direct interaction of NNK with beta-adrenergic, AA-dependent pathways as a novel mechanism of action which may significantly contribute to the high cancer-causing potential of this nitrosamine. Moreover, NNK may also contribute to the development of smoking-related nonneoplastic disease via this mechanism.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Arachidonic Acid/metabolism , Carcinogens/pharmacology , DNA Replication/drug effects , Nitrosamines/pharmacology , Receptors, Adrenergic, beta/genetics , Adenocarcinoma , Animals , Arachidonate 5-Lipoxygenase/metabolism , CHO Cells , Cricetinae , DNA, Neoplasm/drug effects , Humans , Indoles/pharmacology , Lipoxygenase Inhibitors/pharmacology , Lung Neoplasms , Mice , Radioligand Assay , Receptors, Adrenergic, beta/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured
16.
Br J Cancer ; 80(1-2): 175-84, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10389994

ABSTRACT

A model system has been used to test the efficacy of vascular targeting of alpha-particle emitter 213Bi for therapy of small, 'artificial' metastases in mouse lung. Specific monoclonal antibody (mAb) 201 B was used to deliver greater than 30% of the injected dose to lung where tumours had developed due to intravenous injection of cells. Specific 213Bi-mAb 201B treatment of BALB/c mammary carcinoma EMT-6 tumours in lung resulted in a dose-dependent destruction of tumours and an extended lifespan of treated animals relative to controls. Significant reduction of lung tumour burden was noted in animals treated with 0.93 MBq injected dose or as little as 14 Gy absorbed dose to the lung. Animals treated with higher doses (2.6-6.7 MBq) had nearly complete cure of lung tumours but eventually died of lung fibrosis induced by the treatment. Four other tumour cell types were studied: murine Line 1 lung carcinomas in syngeneic BALB/c mice, rat IC-12 tracheal carcinoma growing in severe combined immune deficient (SCID) mice, and two human tumours--epidermoid carcinoma A431 and lung carcinoma A549--growing in SCID mice. In all cases, the number of lung tumour colonies was reduced in animals treated with specific, labelled mAb relative to those in animals treated with control 213Bi MAb or EDTA complexed 213Bi. Tumours treated in immunodeficient SCID mice were partially destroyed or at least retarded in growth, but ultimately regrew and proved fatal, indicating that an intact immune function is necessary for complete cure. The data show that the short-lived alpha-particle emitter 213Bi can be effectively targeted to lung blood vessels and that tumour cells growing in the lung are killed. The mechanism may involve direct killing of tumour cells from alpha-particle irradiation, killing through destruction of blood supply to the tumour, or a combination of the two.


Subject(s)
Bismuth/therapeutic use , Lung Neoplasms/radiotherapy , Radioimmunotherapy , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Animals , Antibodies, Monoclonal , Dose-Response Relationship, Radiation , Lung Neoplasms/blood supply , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, SCID , Microcirculation , Neoplasm Metastasis , Neoplasm Transplantation , Rats , Tumor Cells, Cultured
17.
Acta Orthop Scand ; 69(3): 248-52, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9703397

ABSTRACT

We measured radiographically the polyethylene socket wear in 34 hip prostheses with a 32 mm Al2O3-ceramic head with a rotating bearing and in 37 prostheses with a "fixed" ceramic head. The mean follow-up was 12 years in both groups. The mean annual linear wear of the polyethylene was 0.034 and 0.069 mm, respectively, (Mann-Whitney U-test p < 0.0001) in the "rotation" and the "fixed" group. A rotating bearing between the head and neck in a modular total hip system seems to reduce socket wear as compared to fixed taper junctions.


Subject(s)
Biocompatible Materials , Ceramics , Hip Prosthesis , Materials Testing , Polyethylenes , Prosthesis Failure , Bone Cements , Follow-Up Studies , Hip Joint/diagnostic imaging , Humans , Observer Variation , Prosthesis Design , Radiography , Reoperation , Statistics, Nonparametric
18.
Biochem Pharmacol ; 55(9): 1377-84, 1998 May 01.
Article in English | MEDLINE | ID: mdl-10076528

ABSTRACT

Lung cancer demonstrates a strong etiologic association with smoking. Of the two most common histologic lung cancer types, small cell carcinoma (SCLC) is found almost exclusively in smokers, whereas peripheral adenocarcinoma (PAC) also develops in a significant number of nonsmokers. N'-Nitrosonornicotine (NNN) and 4(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), nicotine-derived nitrosamines, are potent lung carcinogens contained in tobacco products. Because of the structural similarity of NNN and NNK with nicotine, we hypothesized that these carcinogens are ligands for nicotinic acetylcholine receptors (nAChRs). Using cell lines derived from human small cell lung carcinoma and pulmonary adenocarcinoma with the site-selective ligands alpha-bungarotoxin (alpha-BTX) and epibatidine (EB) in receptor binding and cell proliferation assays, we found that SCLC expressed neuronal nicotinic receptors with high affinity to alpha-BTX, whereas PAC cells expressed nicotinic receptors with high affinity to EB. NNK bound with high affinity to alpha-BTX-sensitive nAChRs in SCLC cells, while NNN bound with high affinity to EB sensitive nAChRs in PAC cells. The affinity of each nitrosamine to these receptors was several orders of magnitude greater than that of nicotine. NNK stimulated the proliferation of SCLC cells via this mechanism. Our findings suggest that NNK may contribute to the genesis of SCLC in smokers via chronic stimulation of the alpha BTX-sensitive nAChR-subtype expressed in these cells. Both nitrosamines may also contribute to a host of nicotine-related diseases that are currently thought to be caused by the chronic interaction of nicotine with nAChRs expressed in a large spectrum of mammalian cells.


Subject(s)
Carcinogens/pharmacology , Carcinoma, Small Cell/metabolism , Lung Neoplasms/metabolism , Nicotiana/chemistry , Nitrosamines/pharmacology , Plants, Toxic , Receptors, Nicotinic/metabolism , Adenocarcinoma/metabolism , Binding, Competitive , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bungarotoxins/pharmacology , Carcinogens/pharmacokinetics , Cell Division/drug effects , Humans , Kinetics , Ligands , Nitrosamines/pharmacokinetics , Pyridines/pharmacology , Tumor Cells, Cultured
19.
Mutat Res ; 380(1-2): 13-8, 1997 Oct 31.
Article in English | MEDLINE | ID: mdl-9385385

ABSTRACT

This review is a critical appraisal of our current knowledge on nasal cavity carcinogenesis by nitrosamines. The pathology and pathogenesis of nitrosamine-induced tumors in the nasal cavity of rodents is summarized while controversies on the underlying molecular mechanisms are discussed in more detail. Investigations on the distribution of metabolically competent cell types, the cellular site(s) of nitrosamine metabolism, as well as reports on the cellular distribution and persistence of DNA-adducts strongly suggest that DNA-adducts formed from reactive metabolites are not immediately responsible for the genesis of nasal cavity tumors. A preexisting high proliferative ability has also been suggested as a factor rendering certain cell types more susceptible to the carcinogenic actions of nitrosamines in the nasal cavity. However, this hypothesis has been clearly rejected by more recent investigations. Recent studies have shown that nitrosamines can stimulate the secretion of growth factors via interaction with neurotransmitter receptors in the lungs and that this molecular mechanism is an important factor in determining the histological phenotype of the developing lung tumors. In light of the fact that secretory cells are the main sites of DNA-adduct accumulation and toxic lesions in the nasal cavities of nitrosamine treated rodents, it is suggested that similar mechanisms may mediate the genesis of nitrosamine-induced nasal cavity tumors.


Subject(s)
Nitrosamines/toxicity , Nose Neoplasms/chemically induced , Animals , Cricetinae , DNA Adducts/metabolism , Nitrosamines/metabolism , Organ Specificity , Rats
20.
Chest ; 112(3): 779-84, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9315815

ABSTRACT

Chronic nonneoplastic lung diseases that impair pulmonary oxygenation while increasing the levels of intrapulmonary carbon dioxide (CO2) are a documented risk factor for the development of lung cancer in smokers and nonsmokers. Using established cell lines derived from human small cell lung cancer (SCLC) and non-small cell lung carcinoma, our experiments demonstrated that elevated CO2 concentrations in the range of those found in the diseased lung selectively stimulated the proliferation of SCLC but not adenocarcinoma or squamous cell carcinoma. The proliferative response of SCLC cells involved activation of the mitogen-activated protein kinases ERK-1 and ERK-2, as well as the p70 ribosomal S6 kinase and the stimulation of an autocrine serotonergic loop. Kinase activation was unrelated to changes in intracellular pH. We concluded that CO2 is an important messenger molecule for SCLC which may contribute significantly to the high lung cancer burden observed in individuals with chronic lung disease, by the activation of kinases which play a central role as downstream effectors of many growth factor-stimulated mitogenic pathways.


Subject(s)
Carbon Dioxide/pharmacology , Carcinoma, Small Cell/pathology , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases , Signal Transduction/drug effects , Adenocarcinoma/pathology , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Carbon Dioxide/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Small Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , Chronic Disease , Enzyme Activation/drug effects , Growth Substances/metabolism , Humans , Hydrogen-Ion Concentration , Lung/metabolism , Lung Diseases/metabolism , Lung Neoplasms/etiology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogens/pharmacology , Oxygen Consumption/drug effects , Protein Kinases/drug effects , Protein Serine-Threonine Kinases/drug effects , Protein-Tyrosine Kinases/drug effects , Receptors, Serotonin/drug effects , Ribosomal Protein S6 Kinases , Risk Factors , Serotonin/metabolism , Smoking/adverse effects , Smoking/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...