Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
BMC Med Genomics ; 8: 18, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25944280

ABSTRACT

BACKGROUND: The gene expression profile of cytologically-normal bronchial airway epithelial cells has previously been shown to be altered in patients with lung cancer. Although bronchoscopy is often used for the diagnosis of lung cancer, its sensitivity is imperfect, especially for small and peripheral suspicious lesions. In this study, we derived a gene expression classifier from airway epithelial cells that detects the presence of cancer in current and former smokers undergoing bronchoscopy for suspect lung cancer and evaluated its sensitivity to detect lung cancer among patients from an independent cohort. METHODS: We collected bronchial epithelial cells (BECs) from the mainstem bronchus of 299 current or former smokers (223 cancer-positive and 76 cancer-free subjects) undergoing bronchoscopy for suspected lung cancer in a prospective, multi-center study. RNA from these samples was run on gene expression microarrays for training a gene-expression classifier. A logistic regression model was built to predict cancer status, and the finalized classifier was validated in an independent cohort from a previous study. RESULTS: We found 232 genes whose expression levels in the bronchial airway are associated with lung cancer. We then built a classifier based on the combination of 17 cancer genes, gene expression predictors of smoking status, smoking history, and gender, plus patient age. This classifier had a ROC curve AUC of 0.78 (95% CI, 0.70-0.86) in patients whose bronchoscopy did not lead to a diagnosis of lung cancer (n = 134). In the validation cohort, the classifier had a similar AUC of 0.81 (95% CI, 0.73-0.88) in this same subgroup (n = 118). The classifier performed similarly across a range of mass sizes, cancer histologies and stages. The negative predictive value was 94% (95% CI, 83-99%) in subjects with a non-diagnostic bronchoscopy. CONCLUSION: We developed a gene expression classifier measured in bronchial airway epithelial cells that is able to detect lung cancer in current and former smokers who have undergone bronchoscopy for suspicion of lung cancer. Due to the high NPV of the classifier, it could potentially inform clinical decisions regarding the need for further invasive testing in patients whose bronchoscopy is non diagnostic.


Subject(s)
Bronchi/pathology , Gene Expression Regulation, Neoplastic , Genomics , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Aged , Area Under Curve , Bronchoscopy , Female , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Neoplasms/metabolism , Prospective Studies , ROC Curve , Regression Analysis
2.
N Engl J Med ; 373(3): 243-51, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-25981554

ABSTRACT

BACKGROUND: Bronchoscopy is frequently nondiagnostic in patients with pulmonary lesions suspected to be lung cancer. This often results in additional invasive testing, although many lesions are benign. We sought to validate a bronchial-airway gene-expression classifier that could improve the diagnostic performance of bronchoscopy. METHODS: Current or former smokers undergoing bronchoscopy for suspected lung cancer were enrolled at 28 centers in two multicenter prospective studies (AEGIS-1 and AEGIS-2). A gene-expression classifier was measured in epithelial cells collected from the normal-appearing mainstem bronchus to assess the probability of lung cancer. RESULTS: A total of 639 patients in AEGIS-1 (298 patients) and AEGIS-2 (341 patients) met the criteria for inclusion. A total of 43% of bronchoscopic examinations were nondiagnostic for lung cancer, and invasive procedures were performed after bronchoscopy in 35% of patients with benign lesions. In AEGIS-1, the classifier had an area under the receiver-operating-characteristic curve (AUC) of 0.78 (95% confidence interval [CI], 0.73 to 0.83), a sensitivity of 88% (95% CI, 83 to 92), and a specificity of 47% (95% CI, 37 to 58). In AEGIS-2, the classifier had an AUC of 0.74 (95% CI, 0.68 to 0.80), a sensitivity of 89% (95% CI, 84 to 92), and a specificity of 47% (95% CI, 36 to 59). The combination of the classifier plus bronchoscopy had a sensitivity of 96% (95% CI, 93 to 98) in AEGIS-1 and 98% (95% CI, 96 to 99) in AEGIS-2, independent of lesion size and location. In 101 patients with an intermediate pretest probability of cancer, the negative predictive value of the classifier was 91% (95% CI, 75 to 98) among patients with a nondiagnostic bronchoscopic examination. CONCLUSIONS: The gene-expression classifier improved the diagnostic performance of bronchoscopy for the detection of lung cancer. In intermediate-risk patients with a nondiagnostic bronchoscopic examination, a negative classifier score provides support for a more conservative diagnostic approach. (Funded by Allegro Diagnostics and others; AEGIS-1 and AEGIS-2 ClinicalTrials.gov numbers, NCT01309087 and NCT00746759.).


Subject(s)
Bronchoscopy , Gene Expression Profiling , Gene Expression , Lung Neoplasms/diagnosis , Area Under Curve , Humans , Lung Neoplasms/genetics , Prospective Studies , ROC Curve , Sensitivity and Specificity , Smoking
4.
Cancers (Basel) ; 6(2): 1157-79, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24840047

ABSTRACT

Lung cancer remains the leading cause of cancer-related death in the United States. Cigarette smoking is a well-recognized risk factor for lung cancer, and a sustained elevation of lung cancer risk persists even after smoking cessation. Despite identifiable risk factors, there has been minimal improvement in mortality for patients with lung cancer primarily stemming from diagnosis at a late stage when there are few effective therapeutic options. Early detection of lung cancer and effective screening of high-risk individuals may help improve lung cancer mortality. While low dose computerized tomography (LDCT) screening of high risk smokers has been shown to reduce lung cancer mortality, the high rates of false positives and potential for over-diagnosis have raised questions on how to best implement lung cancer screening. The rapidly evolving field of lung cancer screening and early-detection biomarkers may ultimately improve the ability to diagnose lung cancer in its early stages, identify smokers at highest-risk for this disease, and target chemoprevention strategies. This review aims to provide an overview of the opportunities and challenges related to lung cancer screening, the field of biomarker development for early lung cancer detection, and the future of lung cancer chemoprevention.

6.
Int J Cancer ; 131(12): 2754-62, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22961494

ABSTRACT

Cigarette smoke alters the transcriptome of multiple tissues; those directly exposed to toxic products and those exposed to circulating components and metabolic products of tobacco smoke. In most tissues and organs that have been studied, the smoking transcriptome is characterized by increased expression of antioxidant and xenobiotic genes as well as a wide spectrum of inflammation-related genes, and potential oncogenic genes. Smoking is associated with an increased incidence of cancer in a number of organs both those directly exposed (lungs and airways) and those indirectly exposed (bladder, liver, pancreas). Individual transcriptomic responses vary, based to some degree on as yet to be clarified genetic factors, and likely how and what the individual has smoked. The complexity of individual responses to tobacco exposure and of smoking-related cancers in various organs is beginning to be revealed in transcriptomic and whole genome sequencing studies, of both tumors and cytologically normal appearing cells that have been exposed to cigarette smoke or its products creating a genomic "field of injury." The recent application of next generation sequencing to defining the transcriptome alterations induced by cigarette smoke holds the promise of discovering new approaches to personalized prevention and treatment of smoking-related lung diseases in the future.


Subject(s)
Nicotiana , Smoke/adverse effects , Transcriptome , Humans , Lung Diseases/genetics , Lung Diseases/metabolism , Tissue Distribution
7.
Annu Rev Physiol ; 73: 437-56, 2011.
Article in English | MEDLINE | ID: mdl-21090967

ABSTRACT

Cigarette smoking is responsible for lung cancer and chronic obstructive pulmonary disease (COPD), the leading cause of death from cancer and the second-leading cause of death in the United States. In the United States, 46 million people smoke, with an equal number of former smokers. Moreover, 20-25% of current or former smokers will develop either disease, and smokers with one disease are at increased risk for developing the other. There are no tools for predicting risk of developing either disease; no accepted tools for early diagnosis of potentially curable lung cancer; and no tools for defining molecular pathways or molecular subtypes of these diseases, for predicting rate of progression, or for assessing response to therapy at a biochemical or molecular level. This review discusses current studies and the future potential of measuring global gene expression in epithelial cells that are in the airway field of injury and of using the genomic information derived to begin to answer some of the above questions.


Subject(s)
Carcinoma/chemically induced , Gene Expression/drug effects , Lung Neoplasms/chemically induced , Respiratory Mucosa/drug effects , Smoking/adverse effects , Animals , Carcinoma/epidemiology , Carcinoma/physiopathology , Female , Humans , Lung Neoplasms/epidemiology , Lung Neoplasms/physiopathology , Male , Mice , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/physiopathology , Respiratory Mucosa/metabolism , Smoking/epidemiology , Smoking/physiopathology , United States/epidemiology
8.
Sci Transl Med ; 2(26): 26ra25, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20375364

ABSTRACT

Although only a subset of smokers develop lung cancer, we cannot determine which smokers are at highest risk for cancer development, nor do we know the signaling pathways altered early in the process of tumorigenesis in these individuals. On the basis of the concept that cigarette smoke creates a molecular field of injury throughout the respiratory tract, this study explores oncogenic pathway deregulation in cytologically normal proximal airway epithelial cells of smokers at risk for lung cancer. We observed a significant increase in a genomic signature of phosphatidylinositol 3-kinase (PI3K) pathway activation in the cytologically normal bronchial airway of smokers with lung cancer and smokers with dysplastic lesions, suggesting that PI3K is activated in the proximal airway before tumorigenesis. Further, PI3K activity is decreased in the airway of high-risk smokers who had significant regression of dysplasia after treatment with the chemopreventive agent myo-inositol, and myo-inositol inhibits the PI3K pathway in vitro. These results suggest that deregulation of the PI3K pathway in the bronchial airway epithelium of smokers is an early, measurable, and reversible event in the development of lung cancer and that genomic profiling of these relatively accessible airway cells may enable personalized approaches to chemoprevention and therapy. Our work further suggests that additional lung cancer chemoprevention trials either targeting the PI3K pathway or measuring airway PI3K activation as an intermediate endpoint are warranted.


Subject(s)
Bronchi/enzymology , Bronchi/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Adult , Aged , Bronchi/drug effects , Cohort Studies , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inositol/pharmacology , Lung Neoplasms/genetics , Middle Aged , PTEN Phosphohydrolase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Reproducibility of Results , Smoking/metabolism , Smoking/pathology
9.
PLoS One ; 4(4): e5043, 2009.
Article in English | MEDLINE | ID: mdl-19357784

ABSTRACT

BACKGROUND: Although prior studies have demonstrated a smoking-induced field of molecular injury throughout the lung and airway, the impact of smoking on the airway epithelial proteome and its relationship to smoking-related changes in the airway transcriptome are unclear. METHODOLOGY/PRINCIPAL FINDINGS: Airway epithelial cells were obtained from never (n = 5) and current (n = 5) smokers by brushing the mainstem bronchus. Proteins were separated by one dimensional polyacrylamide gel electrophoresis (1D-PAGE). After in-gel digestion, tryptic peptides were processed via liquid chromatography/ tandem mass spectrometry (LC-MS/MS) and proteins identified. RNA from the same samples was hybridized to HG-U133A microarrays. Protein detection was compared to RNA expression in the current study and a previously published airway dataset. The functional properties of many of the 197 proteins detected in a majority of never smokers were similar to those observed in the never smoker airway transcriptome. LC-MS/MS identified 23 proteins that differed between never and current smokers. Western blotting confirmed the smoking-related changes of PLUNC, P4HB1, and uteroglobin protein levels. Many of the proteins differentially detected between never and current smokers were also altered at the level of gene expression in this cohort and the prior airway transcriptome study. There was a strong association between protein detection and expression of its corresponding transcript within the same sample, with 86% of the proteins detected by LC-MS/MS having a detectable corresponding probeset by microarray in the same sample. Forty-one proteins identified by LC-MS/MS lacked detectable expression of a corresponding transcript and were detected in

Subject(s)
Bronchi/metabolism , Epithelial Cells/physiology , Gene Expression Profiling , Nicotiana , Proteome/analysis , Respiratory Mucosa/cytology , Smoking/adverse effects , Adult , Blotting, Western , Bronchi/cytology , Bronchi/drug effects , Chromatography, Liquid/methods , Electrophoresis, Polyacrylamide Gel/methods , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Glycoproteins/metabolism , Humans , Male , Microarray Analysis , Middle Aged , Molecular Sequence Data , Phosphoproteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Protein Disulfide-Isomerases/metabolism , Respiratory Mucosa/drug effects , Tandem Mass Spectrometry/methods , Uteroglobin/metabolism , Young Adult
10.
Proc Natl Acad Sci U S A ; 106(7): 2319-24, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19168627

ABSTRACT

We have shown that smoking impacts bronchial airway gene expression and that heterogeneity in this response associates with smoking-related disease risk. In this study, we sought to determine whether microRNAs (miRNAs) play a role in regulating the airway gene expression response to smoking. We examined whole-genome miRNA and mRNA expression in bronchial airway epithelium from current and never smokers (n = 20) and found 28 miRNAs to be differentially expressed (P < 0.05) with the majority being down-regulated in smokers. We further identified a number of mRNAs whose expression level is highly inversely correlated with miRNA expression in vivo. Many of these mRNAs contain potential binding sites for the differentially expressed miRNAs in their 3'-untranslated region (UTR) and are themselves affected by smoking. We found that either increasing or decreasing the levels of mir-218 (a miRNA that is strongly affected by smoking) in both primary bronchial epithelial cells and H1299 cells was sufficient to cause a corresponding decrease or increase in the expression of predicted mir-218 mRNA targets, respectively. Further, mir-218 expression is reduced in primary bronchial epithelium exposed to cigarette smoke condensate (CSC), and alteration of mir-218 levels in these cells diminishes the induction of the predicted mir-218 target MAFG in response to CSC. These data indicate that mir-218 levels modulate the airway epithelial gene expression response to cigarette smoke and support a role for miRNAs in regulating host response to environmental toxins.


Subject(s)
Epithelium/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Smoking , Trachea/metabolism , 3' Untranslated Regions , Adult , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Male , MicroRNAs/metabolism , Middle Aged , Risk
11.
BMC Genomics ; 9: 259, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18513428

ABSTRACT

BACKGROUND: Cigarette smoking is a leading cause of preventable death and a significant cause of lung cancer and chronic obstructive pulmonary disease. Prior studies have demonstrated that smoking creates a field of molecular injury throughout the airway epithelium exposed to cigarette smoke. We have previously characterized gene expression in the bronchial epithelium of never smokers and identified the gene expression changes that occur in the mainstem bronchus in response to smoking. In this study, we explored relationships in whole-genome gene expression between extrathorcic (buccal and nasal) and intrathoracic (bronchial) epithelium in healthy current and never smokers. RESULTS: Using genes that have been previously defined as being expressed in the bronchial airway of never smokers (the "normal airway transcriptome"), we found that bronchial and nasal epithelium from non-smokers were most similar in gene expression when compared to other epithelial and nonepithelial tissues, with several antioxidant, detoxification, and structural genes being highly expressed in both the bronchus and nose. Principle component analysis of previously defined smoking-induced genes from the bronchus suggested that smoking had a similar effect on gene expression in nasal epithelium. Gene set enrichment analysis demonstrated that this set of genes was also highly enriched among the genes most altered by smoking in both nasal and buccal epithelial samples. The expression of several detoxification genes was commonly altered by smoking in all three respiratory epithelial tissues, suggesting a common airway-wide response to tobacco exposure. CONCLUSION: Our findings support a relationship between gene expression in extra- and intrathoracic airway epithelial cells and extend the concept of a smoking-induced field of injury to epithelial cells that line the mouth and nose. This relationship could potentially be utilized to develop a non-invasive biomarker for tobacco exposure as well as a non-invasive screening or diagnostic tool providing information about individual susceptibility to smoking-induced lung diseases.


Subject(s)
Bronchi/metabolism , Gene Expression Regulation , Mouth Mucosa/metabolism , Nasal Mucosa/metabolism , Nicotiana , Smoke/adverse effects , Smoking/genetics , Adult , Bronchi/cytology , Case-Control Studies , Epithelial Cells/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genetic Markers , Humans , Male , Middle Aged , Mouth Mucosa/cytology , Nasal Mucosa/cytology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Reproducibility of Results
12.
Cancer Prev Res (Phila) ; 1(6): 396-403, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19138985

ABSTRACT

The concept of field cancerization was first introduced over 6 decades ago in the setting of oral cancer. Later, field cancerization involving histologic and molecular changes of neoplasms and adjacent tissue began to be characterized in smokers with or without lung cancer. Investigators also described a diffuse, nonneoplastic field of molecular injury throughout the respiratory tract that is attributable to cigarette smoking and susceptibility to smoking-induced lung disease. The potential molecular origins of field cancerization and the field of injury following cigarette smoke exposure in lung and airway epithelia are critical to understanding their potential impact on clinical diagnostics and therapeutics for smoking-induced lung disease.


Subject(s)
Lung/pathology , Mouth Mucosa/pathology , Precancerous Conditions/pathology , Respiratory Mucosa/pathology , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Early Detection of Cancer , Environment , Genomics , Humans , Lung/metabolism , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Models, Biological , Mouth Mucosa/metabolism , Precancerous Conditions/diagnosis , Precancerous Conditions/genetics , Respiratory Mucosa/metabolism , Smoking/adverse effects , Smoking/genetics
13.
Genome Biol ; 8(9): R201, 2007.
Article in English | MEDLINE | ID: mdl-17894889

ABSTRACT

BACKGROUND: Tobacco use remains the leading preventable cause of death in the US. The risk of dying from smoking-related diseases remains elevated for former smokers years after quitting. The identification of irreversible effects of tobacco smoke on airway gene expression may provide insights into the causes of this elevated risk. RESULTS: Using oligonucleotide microarrays, we measured gene expression in large airway epithelial cells obtained via bronchoscopy from never, current, and former smokers (n = 104). Linear models identified 175 genes differentially expressed between current and never smokers, and classified these as irreversible (n = 28), slowly reversible (n = 6), or rapidly reversible (n = 139) based on their expression in former smokers. A greater percentage of irreversible and slowly reversible genes were down-regulated by smoking, suggesting possible mechanisms for persistent changes, such as allelic loss at 16q13. Similarities with airway epithelium gene expression changes caused by other environmental exposures suggest that common mechanisms are involved in the response to tobacco smoke. Finally, using irreversible genes, we built a biomarker of ever exposure to tobacco smoke capable of classifying an independent set of former and current smokers with 81% and 100% accuracy, respectively. CONCLUSION: We have categorized smoking-related changes in airway gene expression by their degree of reversibility upon smoking cessation. Our findings provide insights into the mechanisms leading to reversible and persistent effects of tobacco smoke that may explain former smokers increased risk for developing tobacco-induced lung disease and provide novel targets for chemoprophylaxis. Airway gene expression may also serve as a sensitive biomarker to identify individuals with past exposure to tobacco smoke.


Subject(s)
Gene Expression Regulation , Nicotiana/adverse effects , Respiratory System/drug effects , Smoke , Tobacco Smoke Pollution/adverse effects , Trachea/drug effects , Adult , Aged , Biomarkers , Epithelium/drug effects , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Smoking
14.
Ann N Y Acad Sci ; 1098: 389-400, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17435144

ABSTRACT

Optical fiber microarrays have been used to screen saliva from patients with end-stage renal disease (ESRD) to ascertain the efficacy of dialysis. We have successfully identified markers in saliva that correlate with kidney disease. Standard assay chemistries for these markers have been converted to disposable test strips such that patients may one day be able to monitor their clinical status at home. Details of these developments are described. In addition, saliva from asthma and chronic obstructive pulmonary disease (COPD) patients is being screened for useful diagnostic markers. Our goal is to develop a multiplexed assay for these protein and nucleic acid biomarkers for diagnosing the cause and severity of pulmonary exacerbations, enabling more effective treatment to be administered. These results are reported in the second part of this article.


Subject(s)
Microarray Analysis/instrumentation , Saliva/chemistry , Asthma/diagnosis , Asthma/metabolism , Humans , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/metabolism , Oligonucleotide Array Sequence Analysis/instrumentation , Protein Array Analysis/instrumentation , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/metabolism
15.
Nat Med ; 13(3): 361-6, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334370

ABSTRACT

Lung cancer is the leading cause of death from cancer in the US and the world. The high mortality rate (80-85% within 5 years) results, in part, from a lack of effective tools to diagnose the disease at an early stage. Given that cigarette smoke creates a field of injury throughout the airway, we sought to determine if gene expression in histologically normal large-airway epithelial cells obtained at bronchoscopy from smokers with suspicion of lung cancer could be used as a lung cancer biomarker. Using a training set (n = 77) and gene-expression profiles from Affymetrix HG-U133A microarrays, we identified an 80-gene biomarker that distinguishes smokers with and without lung cancer. We tested the biomarker on an independent test set (n = 52), with an accuracy of 83% (80% sensitive, 84% specific), and on an additional validation set independently obtained from five medical centers (n = 35). Our biomarker had approximately 90% sensitivity for stage 1 cancer across all subjects. Combining cytopathology of lower airway cells obtained at bronchoscopy with the biomarker yielded 95% sensitivity and a 95% negative predictive value. These findings indicate that gene expression in cytologically normal large-airway epithelial cells can serve as a lung cancer biomarker, potentially owing to a cancer-specific airway-wide response to cigarette smoke.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Respiratory Mucosa/metabolism , Smoking/adverse effects , Biomarkers/metabolism , Biomarkers, Tumor , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Prospective Studies , Respiratory Mucosa/pathology , Smoking/genetics
16.
Proc Am Thorac Soc ; 3(6): 535-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16921139

ABSTRACT

Both lung cancer and chronic obstructive pulmonary disease (COPD) are associated with cigarette smoking, which, by generating reactive oxidant species, induces a chronic inflammatory state in the lung. Activation, particularly of nuclear factor-kappaB, occurs in both cancer and COPD, and expression of a number of genes is altered in both diseases. In lung cancer, DNA damage, lack of DNA repair, and genomic instability predominate, whereas matrix degradation, lack of repair, and an intense immune response predominate in COPD. The reasons for the different responses to a common inflammatory response induced by smoking remain to be determined, but likely lie in genetic polymorphisms in genes that regulate genome integrity in cancer and that regulate the immune response to tissue destruction in COPD.


Subject(s)
Lung Neoplasms/etiology , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , Immunity, Cellular , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mutation , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology
17.
Nucleic Acids Res ; 33(Database issue): D573-9, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15608264

ABSTRACT

The SIEGE (Smoking Induced Epithelial Gene Expression) database is a clinical resource for compiling and analyzing gene expression data from epithelial cells of the human intra-thoracic airway. This database supports a translational research study whose goal is to profile the changes in airway gene expression that are induced by cigarette smoke. RNA is isolated from airway epithelium obtained at bronchoscopy from current-, former- and never-smoker subjects, and hybridized to Affymetrix HG-U133A Genechips, which measure the level of expression of approximately 22,500 human transcripts. The microarray data generated along with relevant patient information is uploaded to SIEGE by study administrators using the database's web interface, found at http://pulm.bumc.bu.edu/siegeDB. PERL-coded scripts integrated with SIEGE perform various quality control functions including the processing, filtering and formatting of stored data. The R statistical package is used to import database expression values and execute a number of statistical analyses including t-tests, correlation coefficients and hierarchical clustering. Values from all statistical analyses can be queried through CGI-based tools and web forms found on the 'Search' section of the database website. Query results are embedded with graphical capabilities as well as with links to other databases containing valuable gene resources, including Entrez Gene, GO, Biocarta, GeneCards, dbSNP and the NCBI Map Viewer.


Subject(s)
Databases, Genetic , Gene Expression Profiling , Respiratory Mucosa/metabolism , Smoking , Humans , Oligonucleotide Array Sequence Analysis , User-Computer Interface
18.
Am J Respir Cell Mol Biol ; 31(6): 601-10, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15374838

ABSTRACT

The mechanism by which inhaled smoke causes the anatomic lesions and physiologic impairment of chronic obstructive pulmonary disease remains unknown. We used high-density microarrays to measure gene expression in severely emphysematous lung tissue removed from smokers at lung volume reduction surgery (LVRS) and normal or mildly emphysematous lung tissue from smokers undergoing resection of pulmonary nodules. Class prediction algorithms identified 102 genes that accurately distinguished severe emphysema from non-/mildly emphysematous lung tissue. We also defined a number of genes whose expression levels correlated strongly with lung diffusion capacity for carbon monoxide and/or forced expiratory volume at 1 s. Genes related to oxidative stress, extracellular matrix synthesis, and inflammation were increased in severe emphysema, whereas expression of endothelium-related genes was decreased. To identify candidate genes that might be causally involved in the pathogenesis of emphysema, we linked gene expression profiles to chromosomal regions previously associated with chronic obstructive pulmonary disease in genome-wide linkage analyses. Unsupervised hierarchical clustering of the LVRS samples revealed distinct molecular subclasses of severe emphysema, with body mass index as the only clinical variable that differed between the groups. Class prediction models established a set of genes that predicted functional outcome at 6 mo after LVRS. Our findings suggest that the gene expression profiles from human emphysematous lung tissue may provide insight into pathogenesis, uncover novel molecular subclasses of disease, predict response to LVRS, and identify targets for therapeutic intervention.


Subject(s)
Gene Expression Profiling , Lung/metabolism , Lung/pathology , Pulmonary Emphysema/genetics , Smoking/genetics , Aged , Case-Control Studies , Chromosomes, Human/genetics , Female , Humans , Lod Score , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pulmonary Emphysema/surgery , RNA, Messenger/analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
19.
Proc Natl Acad Sci U S A ; 101(27): 10143-8, 2004 Jul 06.
Article in English | MEDLINE | ID: mdl-15210990

ABSTRACT

Cigarette smoke is the major cause of lung cancer, the leading cause of cancer death, and of chronic obstructive pulmonary disease, the fourth leading cause of death in the United States. Using high-density gene expression arrays, we describe genes that are normally expressed in a subset of human airway epithelial cells obtained at bronchoscopy (the airway transcriptome), define how cigarette smoking alters the transcriptome, and detail the effects of variables, such as cumulative exposure, age, sex, and race, on cigarette smoke-induced changes in gene expression. We also determine which changes in gene expression are and are not reversible when smoking is discontinued. The persistent altered expression of a subset of genes in former smokers may explain the risk these individuals have for developing lung cancer long after they have discontinued smoking. The use of gene expression profiling to explore the normal biology of a specific subset of cells within a complex organ across a broad spectrum of healthy individuals and to define the reversible and irreversible genetic effects of cigarette smoke on human airway epithelial cells has not been previously reported.


Subject(s)
Bronchi/metabolism , Gene Expression Profiling , Smoking/metabolism , Epithelial Cells/metabolism , Humans , Smoking Cessation , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL