Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article En | MEDLINE | ID: mdl-33903236

Molecular events that drive the development of precancerous lesions in the bronchial epithelium, which are precursors of lung squamous cell carcinoma (LUSC), are poorly understood. We demonstrate that disruption of epithelial cellular polarity, via the conditional deletion of the apical determinant Crumbs3 (Crb3), initiates and sustains precancerous airway pathology. The loss of Crb3 in adult luminal airway epithelium promotes the uncontrolled activation of the transcriptional regulators YAP and TAZ, which stimulate intrinsic signals that promote epithelial cell plasticity and paracrine signals that induce basal-like cell growth. We show that aberrant polarity and YAP/TAZ-regulated gene expression associates with human bronchial precancer pathology and disease progression. Analyses of YAP/TAZ-regulated genes further identified the ERBB receptor ligand Neuregulin-1 (NRG1) as a key transcriptional target and therapeutic targeting of ERBB receptors as a means of preventing and treating precancerous cell growth. Our observations offer important molecular insight into the etiology of LUSC and provides directions for potential interception strategies of lung cancer.


Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Membrane Glycoproteins/genetics , Neuregulin-1/genetics , Precancerous Conditions/genetics , YAP-Signaling Proteins/genetics , Carcinoma, Squamous Cell/pathology , Cell Polarity/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Epithelium/pathology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Signal Transduction/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics
2.
Chest ; 156(4): 764-773, 2019 10.
Article En | MEDLINE | ID: mdl-31233743

BACKGROUND: Although e-cigarette (ECIG) use has increased in the United States, their potential health effects remain uncertain. Understanding the effects of tobacco cigarette (TCIG) smoke on bronchial airway epithelial gene expression have previously provided insights into tobacco-related disease pathogenesis. Identifying the impact of ECIGs on airway gene expression could provide insights into their potential long-term health effects. We sought to compare the bronchial airway gene-expression profiles of former TCIG smokers now using ECIGs with the profiles of former and current TCIG smokers. METHODS: We performed gene-expression profiling of bronchial epithelial cells collected from current TCIG smokers (n = 9), current ECIG users who are former TCIG smokers (n = 15), and former TCIG smokers (n = 21). We then compared our findings with previous studies of the effects of TCIG use on bronchial epithelium, as well an in vitro model of ECIG exposure. RESULTS: Among 3,165 genes whose expression varied between the three study groups (q < 0.05), we identified 468 genes altered in ECIG users relative to former smokers (P < .05). Seventy-nine of these genes were up- or down-regulated concordantly among ECIG and TCIG users. We did not detect ECIG-associated gene-expression changes in known pathways associated with TCIG usage. Genes downregulated in ECIG users are enriched among the genes most downregulated by exposure of airway epithelium to ECIG vapor in vitro. CONCLUSIONS: ECIGs induce both distinct and shared patterns of gene expression relative to TCIGs in the bronchial airway epithelium. The concordance of the genes altered in ECIG users and in the in vitro study suggests that genes altered in ECIG users are likely to be changed as the direct effect of ECIG exposure.


Bronchi/cytology , Electronic Nicotine Delivery Systems , Epithelial Cells , Gene Expression Regulation , Smoking/genetics , Adult , Cigarette Smoking/genetics , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Smoking/adverse effects
3.
Nat Commun ; 10(1): 1856, 2019 04 23.
Article En | MEDLINE | ID: mdl-31015447

Bronchial premalignant lesions (PMLs) are precursors of lung squamous cell carcinoma, but have variable outcome, and we lack tools to identify and treat PMLs at risk for progression to cancer. Here we report the identification of four molecular subtypes of PMLs with distinct differences in epithelial and immune processes based on RNA-Seq profiling of endobronchial biopsies from high-risk smokers. The Proliferative subtype is enriched with bronchial dysplasia and exhibits up-regulation of metabolic and cell cycle pathways. A Proliferative subtype-associated gene signature identifies subjects with Proliferative PMLs from normal-appearing uninvolved large airway brushings with high specificity. In progressive/persistent Proliferative lesions expression of interferon signaling and antigen processing/presentation pathways decrease and immunofluorescence indicates a depletion of innate and adaptive immune cells compared with regressive lesions. Molecular biomarkers measured in PMLs or the uninvolved airway can enhance histopathological grading and suggest immunoprevention strategies for intercepting the progression of PMLs to lung cancer.


Biomarkers, Tumor/genetics , Carcinoma, Bronchogenic/pathology , Gene Expression Regulation, Neoplastic/immunology , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/immunology , Biopsy , Bronchi/diagnostic imaging , Bronchi/immunology , Bronchi/pathology , Bronchoscopy , Carcinoma, Bronchogenic/genetics , Carcinoma, Bronchogenic/immunology , Carcinoma, Bronchogenic/prevention & control , Cohort Studies , Datasets as Topic , Disease Progression , Early Detection of Cancer/methods , Gene Expression Profiling , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Mass Screening/methods , Middle Aged , Precancerous Conditions/diagnostic imaging , Precancerous Conditions/genetics , Precancerous Conditions/immunology , RNA, Messenger/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/diagnostic imaging , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Sequence Analysis, RNA , T-Lymphocytes/immunology , Tomography, X-Ray Computed , Up-Regulation
4.
Front Genet ; 10: 150, 2019.
Article En | MEDLINE | ID: mdl-30891063

The need to reduce per sample cost of RNA-seq profiling for scalable data generation has led to the emergence of highly multiplexed RNA-seq. These technologies utilize barcoding of cDNA sequences in order to combine multiple samples into a single sequencing lane to be separated during data processing. In this study, we report the performance of one such technique denoted as sparse full length sequencing (SFL), a ribosomal RNA depletion-based RNA sequencing approach that allows for the simultaneous sequencing of 96 samples and higher. We offer comparisons to well established single-sample techniques, including: full coverage Poly-A capture RNA-seq, microarrays, as well as another low-cost highly multiplexed technique known as 3' digital gene expression (3'DGE). Data was generated for a set of exposure experiments on immortalized human lung epithelial (AALE) cells in a two-by-two study design, in which samples received both genetic and chemical perturbations of known oncogenes/tumor suppressors and lung carcinogens. SFL demonstrated improved performance over 3'DGE in terms of coverage, power to detect differential gene expression, and biological recapitulation of patterns of differential gene expression from in vivo lung cancer mutation signatures.

5.
Toxicol Sci ; 155(1): 248-257, 2017 01.
Article En | MEDLINE | ID: mdl-27701119

Little evidence is available regarding the physiological effects of exposure to electronic cigarette (ECIG) aerosol. We sought to determine the molecular impact of ECIG aerosol exposure in human bronchial epithelial cells (HBECs). Gene-expression profiling was conducted in primary grown at air liquid interface and exposed to 1 of 4 different ECIG aerosols, traditional tobacco cigarette (TCIG) smoke, or clean air. Findings were validated experimentally with quantitative polymerase chain reaction and a reactive oxygen species immunoassay. Using gene set enrichment analysis, signatures of in vitro ECIG exposure were compared with those generated from bronchial epithelial brushings of current TCIG smokers and former TCIG smokers currently using ECIGs. We found 546 genes differentially expressed across the ECIG, TCIG, and air-exposed groups of HBECs (ANOVA; FDR q < .05; fold change > 1.5). A subset of these changes were shared between TCIG- and ECIG-exposed HBECs. ECIG exposure induced genes involved in oxidative and xenobiotic stress pathways and increased a marker of reactive oxygen species production in a dose-dependent manner. ECIG exposure decreased expression of genes involved in cilia assembly and movement. Furthermore, gene-expression differences observed in vitro were concordant with differences observed in airway epithelium collected from ECIG users (q < .01). In summary, our data suggest that ECIG aerosol can induce gene-expression changes in bronchial airway epithelium in vitro, some of which are shared with TCIG smoke. These changes were generally less pronounced than the effects of TCIG exposure and were more pronounced in ECIG products containing nicotine than those without nicotine. Our data further suggest that the gene-expression alterations seen with the in vitro exposure system reflects the physiological effects experienced in vivo by ECIG users.


Aerosols , Bronchi/drug effects , Electronic Nicotine Delivery Systems , Epithelium/drug effects , Humans , Real-Time Polymerase Chain Reaction
6.
Am J Respir Crit Care Med ; 194(8): 948-960, 2016 10 15.
Article En | MEDLINE | ID: mdl-27104832

RATIONALE: Despite shared environmental exposures, idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease are usually studied in isolation, and the presence of shared molecular mechanisms is unknown. OBJECTIVES: We applied an integrative genomic approach to identify convergent transcriptomic pathways in emphysema and IPF. METHODS: We defined the transcriptional repertoire of chronic obstructive pulmonary disease, IPF, or normal histology lungs using RNA-seq (n = 87). MEASUREMENTS AND MAIN RESULTS: Genes increased in both emphysema and IPF relative to control were enriched for the p53/hypoxia pathway, a finding confirmed in an independent cohort using both gene expression arrays and the nCounter Analysis System (n = 193). Immunohistochemistry confirmed overexpression of HIF1A, MDM2, and NFKBIB members of this pathway in tissues from patients with emphysema or IPF. Using reads aligned across splice junctions, we determined that alternative splicing of p53/hypoxia pathway-associated molecules NUMB and PDGFA occurred more frequently in IPF or emphysema compared with control and validated these findings by quantitative polymerase chain reaction and the nCounter Analysis System on an independent sample set (n = 193). Finally, by integrating parallel microRNA and mRNA-Seq data on the same samples, we identified MIR96 as a key novel regulatory hub in the p53/hypoxia gene-expression network and confirmed that modulation of MIR96 in vitro recapitulates the disease-associated gene-expression network. CONCLUSIONS: Our results suggest convergent transcriptional regulatory hubs in diseases as varied phenotypically as chronic obstructive pulmonary disease and IPF and suggest that these hubs may represent shared key responses of the lung to environmental stresses.


Gene Regulatory Networks/genetics , Idiopathic Pulmonary Fibrosis/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Adult , Emphysema/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , I-kappa B Proteins/metabolism , Male , Membrane Proteins/metabolism , Middle Aged , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism
7.
Proc Natl Acad Sci U S A ; 110(47): 18946-51, 2013 Nov 19.
Article En | MEDLINE | ID: mdl-24158479

Smoking is a significant risk factor for lung cancer, the leading cause of cancer-related deaths worldwide. Although microRNAs are regulators of many airway gene-expression changes induced by smoking, their role in modulating changes associated with lung cancer in these cells remains unknown. Here, we use next-generation sequencing of small RNAs in the airway to identify microRNA 4423 (miR-4423) as a primate-specific microRNA associated with lung cancer and expressed primarily in mucociliary epithelium. The endogenous expression of miR-4423 increases as bronchial epithelial cells undergo differentiation into mucociliary epithelium in vitro, and its overexpression during this process causes an increase in the number of ciliated cells. Furthermore, expression of miR-4423 is reduced in most lung tumors and in cytologically normal epithelium of the mainstem bronchus of smokers with lung cancer. In addition, ectopic expression of miR-4423 in a subset of lung cancer cell lines reduces their anchorage-independent growth and significantly decreases the size of the tumors formed in a mouse xenograft model. Consistent with these phenotypes, overexpression of miR-4423 induces a differentiated-like pattern of airway epithelium gene expression and reverses the expression of many genes that are altered in lung cancer. Together, our results indicate that miR-4423 is a regulator of airway epithelium differentiation and that the abrogation of its function contributes to lung carcinogenesis.


Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Cell Differentiation/physiology , Lung Neoplasms/diagnosis , MicroRNAs/metabolism , Respiratory Mucosa/cytology , Animals , Biomarkers, Tumor/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Immunohistochemistry , In Situ Hybridization , Lung Neoplasms/genetics , Mice , MicroRNAs/genetics , Microarray Analysis , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/metabolism
8.
Am J Respir Crit Care Med ; 187(9): 933-42, 2013 May 01.
Article En | MEDLINE | ID: mdl-23471465

RATIONALE: Molecular phenotyping of chronic obstructive pulmonary disease (COPD) has been impeded in part by the difficulty in obtaining lung tissue samples from individuals with impaired lung function. OBJECTIVES: We sought to determine whether COPD-associated processes are reflected in gene expression profiles of bronchial airway epithelial cells obtained by bronchoscopy. METHODS: Gene expression profiling of bronchial brushings obtained from 238 current and former smokers with and without COPD was performed using Affymetrix Human Gene 1.0 ST Arrays. MEASUREMENTS AND MAIN RESULTS: We identified 98 genes whose expression levels were associated with COPD status, FEV1% predicted, and FEV1/FVC. In silico analysis identified activating transcription factor 4 (ATF4) as a potential transcriptional regulator of genes with COPD-associated airway expression, and ATF4 overexpression in airway epithelial cells in vitro recapitulates COPD-associated gene expression changes. Genes with COPD-associated expression in the bronchial airway epithelium had similarly altered expression profiles in prior studies performed on small-airway epithelium and lung parenchyma, suggesting that transcriptomic alterations in the bronchial airway epithelium reflect molecular events found at more distal sites of disease activity. Many of the airway COPD-associated gene expression changes revert toward baseline after therapy with the inhaled corticosteroid fluticasone in independent cohorts. CONCLUSIONS: Our findings demonstrate a molecular field of injury throughout the bronchial airway of active and former smokers with COPD that may be driven in part by ATF4 and is modifiable with therapy. Bronchial airway epithelium may ultimately serve as a relatively accessible tissue in which to measure biomarkers of disease activity for guiding clinical management of COPD.


Activating Transcription Factor 4/genetics , Bronchi/metabolism , Epithelial Cells/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Smoking/adverse effects , Transcriptome/physiology , Aged , Analysis of Variance , Androstadienes , Bronchi/drug effects , Bronchodilator Agents/pharmacology , Bronchoscopy , Epithelial Cells/drug effects , Female , Fluticasone , Humans , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Real-Time Polymerase Chain Reaction , Respiratory Function Tests , Transcriptome/drug effects
9.
Mutat Res ; 717(1-2): 32-7, 2011 Dec 01.
Article En | MEDLINE | ID: mdl-21549133

miRNAs are a class of small, noncoding RNAs averaging 22 nucleotides in length that down-regulate gene expression by complimentary binding to the 3' UTR of target genes. A growing body of research suggests that these small RNA species play significant roles in modulating the cellular response to a variety of types of stress. In this review, we summarize the available literature regarding the general response of miRNA to cellular stress, and then specifically focus on the miRNA response to inhaled toxins. These miRNA responses to inhaled toxins appear to be recapitulated in lung carcinogenesis, opening the possibility that modulation of the miRNA response could be a novel strategy for chemoprevention.


Biomarkers/metabolism , Environmental Pollutants/toxicity , MicroRNAs/metabolism , Cell Transformation, Neoplastic , Humans , Inhalation Exposure , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MicroRNAs/genetics , Stress, Physiological , Tobacco Smoke Pollution/adverse effects
10.
Cell Cycle ; 9(9): 1809-18, 2010 May.
Article En | MEDLINE | ID: mdl-20404570

Reduced expression of miR-129 has been reported in multiple tumor cell lines and in primary tumors including medulloblastoma, undifferentiated gastric cancers, lung adenocarcinoma, endometrial cancer and colorectal carcinoma. There is also recent evidence of an anti-proliferative activity of miR-129 in tumor cell lines. Still, little is known about how miR-129 regulates cell proliferation. Here we found that lentiviral-mediated overexpression of miR-129 in mouse lung epithelial cells (E10 cells) results in significant G(1) phase arrest that eventually leads to cell death. miR-129 induce G(1) phase arrest in multiple human lung adenocarcinoma cell lines, suggesting miR-129 targeting of G(1)/S phase-specific regulators. Interestingly, we show that Cdk6, a kinase involved in G(1)-S transition, is a direct target of miR-129. We also found the downregulation of three other cell cycle-related novel targets of miR-129, including Erk1, Erk2 and protein kinase C epsilon (Prkce). We further show that among these targets, only Cdk6 is functionally relevant. Restoring expression of Cdk6, but not Prkce partially rescues the cell growth arrest and cell death phenotype that results from miR-129 overexpression. Together, our data indicate that miR-129 plays an important role in regulating cell proliferation by downregulation of Cdk6.


Cyclin-Dependent Kinase 6/metabolism , MicroRNAs/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase 6/genetics , Down-Regulation , G1 Phase , Gene Expression Regulation, Neoplastic , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-epsilon/metabolism , S Phase
11.
Proc Natl Acad Sci U S A ; 106(7): 2319-24, 2009 Feb 17.
Article En | MEDLINE | ID: mdl-19168627

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.


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
...