Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Cancer Res ; 67(10): 4638-47, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17510389

ABSTRACT

Nicotine and its derivatives, by binding to nicotinic acetylcholine receptors (nAChR) on bronchial epithelial cells, can regulate cellular proliferation and apoptosis via activating the Akt pathway. Delineation of nAChR subtypes in non-small-cell lung cancers (NSCLC) may provide information for prevention or therapeutic targeting. Expression of nAChR subunit genes in 66 resected primary NSCLCs, 7 histologically non-involved lung tissues, 13 NSCLC cell lines, and 6 human bronchial epithelial cell lines (HBEC) was analyzed with quantitative PCR and microarray analysis. Five nonmalignant HBECs were exposed to nicotine in vitro to study the variation of nAChR subunit gene expression with nicotine exposure and removal. NSCLCs from nonsmokers showed higher expression of nAChR alpha6 (P < 0.001) and beta3 (P = 0.007) subunit genes than those from smokers, adjusted for gender. In addition, nAChR alpha4 (P < 0.001) and beta4 (P = 0.029) subunit gene expression showed significant difference between NSCLCs and normal lung. Using Affymetrix GeneChip U133 Sets, 65 differentially expressed genes associated with NSCLC nonsmoking nAChR alpha6beta3 phenotype were identified, which gave high sensitivity and specificity of prediction. nAChR alpha1, alpha5, and alpha7 showed significant reversible changes in expression levels in HBECs upon nicotine exposure. We conclude that between NSCLCs from smokers and nonsmokers, different nAChR subunit gene expression patterns were found, and a 65-gene expression signature was associated with nonsmoking nAChR alpha6beta3 expression. Finally, nicotine exposure in HBECs resulted in reversible differences in nAChR subunit gene expression. These results further implicate nicotine in bronchial carcinogenesis and suggest targeting nAChRs for prevention and therapy in lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Receptors, Nicotinic/genetics , Smoking/genetics , Adult , Aged , Aged, 80 and over , Bronchi/cytology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Gene Expression , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Nicotine/pharmacology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Nicotinic/biosynthesis , Smoking/adverse effects , Smoking/metabolism
2.
Curr Biol ; 15(20): 1820-6, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16243028

ABSTRACT

Activity-prompted dendritic remodeling leads to calcium-influx-dependent activation of signaling pathways within minutes and gene transcription within hours. However, dendrite growth continues for days and requires extension and stabilization of the cytoskeleton in nascent processes. In addition to binding microtubules, microtubule-associated proteins (MAPs) associate with the actin cytoskeleton, anchor ion channels and signaling complexes, and modulate synaptic growth. MAP2 is predominantly dendritic. MAP1B is at postsynaptic densities (PSD) and modulates ion channel activity, in addition to affecting axon growth. Less is known about MAP1A, but it is also enriched in dendrites at input locations, including PSDs where MAP1A associates with channel complexes and the calcium sensor caldendrin. MAP1A rescued hearing loss in tubby mice. Here we show that MAP1A becomes enriched in dendrites concurrently with dendritic branching and synapse formation in the developing brain; that synaptic activity is required for establishing mature MAP1A expression levels; and that MAP1A expression is required for activity-dependent growth, branching, and stabilization of the dendritic arbor.


Subject(s)
Dendrites/physiology , Gene Expression , Hippocampus/cytology , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Animals , Hippocampus/growth & development , Immunoblotting , Immunohistochemistry , Microscopy, Fluorescence , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley
3.
Cancer Res ; 64(24): 9027-34, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15604268

ABSTRACT

By expressing two genes (hTERT and Cdk4), we have developed a method to reproducibly generate continuously replicating human bronchial epithelial cell (HBEC) lines that provide a novel resource to study the molecular pathogenesis of lung cancer and the differentiation of bronchial epithelial cells. Twelve human bronchial epithelial biopsy specimens obtained from persons with and without lung cancer were placed into short-term culture and serially transfected with retroviral constructs containing cyclin-dependent kinase (Cdk) 4 and human telomerase reverse transcriptase (hTERT), resulting in continuously growing cultures. The order of introduction of Cdk4 and hTERT did not appear to be important; however, transfection of either gene alone did not result in immortalization. Although they could be cloned, the immortalized bronchial cells did not form colonies in soft agar or tumors in nude mice. The immortalized HBECs have epithelial morphology; express epithelial markers cytokeratins 7, 14, 17, and 19, the stem cell marker p63, and high levels of p16(INK4a); and have an intact p53 checkpoint pathway. Cytogenetic analysis and array comparative genomic hybridization profiling show immortalized HBECs to have duplication of parts of chromosomes 5 and 20. Microarray gene expression profiling demonstrates that the Cdk4/hTERT-immortalized bronchial cell lines clustered together and with nonimmortalized bronchial cells, distinct from lung cancer cell lines. We also immortalized several parental cultures with viral oncoproteins human papilloma virus type 16 E6/E7 with and without hTERT, and these cells exhibited loss of the p53 checkpoint and significantly different gene expression profiles compared with Cdk4/hTERT-immortalized HBECs. These HBEC lines are a valuable new tool for studying of the pathogenesis of lung cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lung/metabolism , Lung/physiology , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinases/biosynthesis , Cyclin-Dependent Kinases/genetics , DNA-Binding Proteins , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Immunoblotting , Karyotyping , Lung/cytology , Nucleic Acid Hybridization , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Telomerase/biosynthesis , Telomerase/genetics , Telomere/genetics , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factors , Transfection , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...