Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Cancer ; 144(5): 1104-1114, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30152518

ABSTRACT

Somatostatin receptor 2 (SSTR2) is overexpressed in a majority of neuroendocrine neoplasms, including small-cell lung carcinomas (SCLCs). SSTR2 was previously considered an inhibitory receptor on cell growth, but its agonists had poor clinical responses in multiple clinical trials. The role of this receptor as a potential therapeutic target in lung cancer merits further investigation. We evaluated the expression of SSTR2 in a cohort of 96 primary tumors from patients with SCLC and found 48% expressed SSTR2. Correlation analysis in both CCLE and an SCLC RNAseq cohort confirmed high-level expression and identified an association between NEUROD1 and SSTR2. There was a significant association with SSTR2 expression profile and poor clinical outcome. We tested whether SSTR2 expression might contribute to tumor progression through activation of downstream signaling pathways, using in vitro and in vivo systems and downregulated SSTR2 expression in lung cancer cells by shRNA. SSTR2 downregulation led to increased apoptosis and dramatically decreased tumor growth in vitro and in vivo in multiple cell lines with decreased AMPKα phosphorylation and increased oxidative metabolism. These results demonstrate a role for SSTR2 signaling in SCLC and suggest that SSTR2 is a poor prognostic biomarker in SCLC and potential future therapeutic signaling target.


Subject(s)
Cell Proliferation/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Receptors, Somatostatin/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Disease Progression , Down-Regulation/genetics , Humans , Mice , Mice, Nude , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Signal Transduction/genetics
2.
Oncogene ; 37(36): 5007-5019, 2018 09.
Article in English | MEDLINE | ID: mdl-29789716

ABSTRACT

Many tumors increase uptake and dependence on glucose, cystine or glutamine. These basic observations on cancer cell metabolism have opened multiple new diagnostic and therapeutic avenues in cancer research. Recent studies demonstrated that smoking could induce the expression of xCT (SLC7A11) in oral cancer cells, suggesting that overexpression of xCT may support lung tumor progression. We hypothesized that overexpression of xCT occurs in lung cancer cells to satisfy the metabolic requirements for growth and survival. Our results demonstrated that 1) xCT was highly expressed at the cytoplasmic membrane in non-small cell lung cancer (NSCLC), 2) the expression of xCT was correlated with advanced stage and predicted a worse 5-year survival, 3) targeting xCT transport activity in xCT overexpressing NSCLC cells with sulfasalazine decreased cell proliferation and invasion in vitro and in vivo and 4) increased dependence on glutamine was observed in xCT overexpressed normal airway epithelial cells. These results suggested that xCT regulate metabolic requirements during lung cancer progression and be a potential therapeutic target in NSCLC.


Subject(s)
Amino Acid Transport System y+/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , 3T3 Cells , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Cystine/metabolism , Cytoplasm/metabolism , Disease Progression , Female , Glutamine/metabolism , Humans , Male , Mice , Middle Aged
3.
J Proteome Res ; 16(9): 3266-3276, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28731711

ABSTRACT

We hypothesized that distinct protein expression features of benign and malignant pulmonary nodules may reveal novel candidate biomarkers for the early detection of lung cancer. We performed proteome profiling by liquid chromatography-tandem mass spectrometry to characterize 34 resected benign lung nodules, 24 untreated lung adenocarcinomas (ADCs), and biopsies of bronchial epithelium. Group comparisons identified 65 proteins that differentiate nodules from ADCs and normal bronchial epithelium and 66 proteins that differentiate ADCs from nodules and normal bronchial epithelium. We developed a multiplexed parallel reaction monitoring (PRM) assay to quantify a subset of 43 of these candidate biomarkers in an independent cohort of 20 benign nodules, 21 ADCs, and 20 normal bronchial biopsies. PRM analyses confirmed significant nodule-specific abundance of 10 proteins including ALOX5, ALOX5AP, CCL19, CILP1, COL5A2, ITGB2, ITGAX, PTPRE, S100A12, and SLC2A3 and significant ADC-specific abundance of CEACAM6, CRABP2, LAD1, PLOD2, and TMEM110-MUSTN1. Immunohistochemistry analyses for seven selected proteins performed on an independent set of tissue microarrays confirmed nodule-specific expression of ALOX5, ALOX5AP, ITGAX, and SLC2A3 and cancer-specific expression of CEACAM6. These studies illustrate the value of global and targeted proteomics in a systematic process to identify and qualify candidate biomarkers for noninvasive molecular diagnosis of lung cancer.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Neoplasm Proteins/genetics , Solitary Pulmonary Nodule/diagnosis , 5-Lipoxygenase-Activating Proteins/genetics , 5-Lipoxygenase-Activating Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Biomarkers, Tumor/metabolism , CD11 Antigens/genetics , CD11 Antigens/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Diagnosis, Differential , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/metabolism , Proteomics/methods , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Solitary Pulmonary Nodule/genetics , Solitary Pulmonary Nodule/metabolism , Solitary Pulmonary Nodule/pathology , Tandem Mass Spectrometry , Tissue Array Analysis , Transcriptome
4.
JCI Insight ; 1(19): e88814, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27882349

ABSTRACT

The molecular determinants of lung cancer risk remain largely unknown. Airway epithelial cells are prone to assault by risk factors and are considered to be the primary cell type involved in the field of cancerization. To investigate risk-associated changes in the bronchial epithelium proteome that may offer new insights into the molecular pathogenesis of lung cancer, proteins were identified in the airway epithelial cells of bronchial brushing specimens from risk-stratified individuals by shotgun proteomics. Differential expression of selected proteins was validated by parallel reaction monitoring mass spectrometry in an independent set of individual bronchial brushings. We identified 2,869 proteins, of which 312 proteins demonstrated a trend in expression. Pathway analysis revealed enrichment of carbohydrate metabolic enzymes in high-risk individuals. Glucose consumption and lactate production were increased in human bronchial epithelial BEAS2B cells treated with cigarette smoke condensate for 7 months. Increased lipid biosynthetic capacity and net reductive carboxylation were revealed by metabolic flux analyses of [U-13C5] glutamine in this in vitro model, suggesting profound metabolic reprogramming in the airway epithelium of high-risk individuals. These results provide a rationale for the development of potentially new chemopreventive strategies and selection of patients for surveillance programs.


Subject(s)
Epithelial Cells/metabolism , Proteome/analysis , Respiratory Mucosa/pathology , Smoke/adverse effects , Bronchi , Cell Line , Gene Expression Profiling , Humans , Lipid Metabolism , Lung Neoplasms/metabolism , Metabolomics , Respiratory Mucosa/cytology , Smoking
5.
J Immunol Methods ; 438: 51-58, 2016 11.
Article in English | MEDLINE | ID: mdl-27594594

ABSTRACT

T and B lymphocytes undergo metabolic re-programming upon activation that is essential to allow bioenergetics, cell survival, and intermediates for cell proliferation and function. To support changes in the activity of signaling pathways and to provide sufficient and necessary intracellular metabolites, uptake of extracellular nutrients increases sharply with metabolic re-programming. One result of increased metabolic activity can be reactive oxygen species (ROS), which can be toxic when accumulated in excess. Uptake of cystine allows accumulation of cysteine that is necessary for glutathione synthesis and ROS detoxification. Cystine uptake is required for T cell activation and function but measurements based on radioactive labeling do not allow analysis on single cell level. Here we show the critical role for cystine uptake in T cells using a method for measurement of cystine uptake using a novel CystineFITC probe. T cell receptor stimulation lead to upregulation of the cystine transporter xCT (SLC7a11) and increased cystine uptake in CD4+ and CD8+ human T cells. Similarly, lipopolysaccharide stimulation increased cystine uptake in human B cells. The CystineFITC probe was not toxic and could be metabolized to prevent cystine starvation induced cell death. Furthermore, blockade of xCT or competition with natural cystine decreased uptake of CystineFITC. CystineFITC is thus a versatile tool that allows measurement of cystine uptake on single cell level and shows the critical role for cystine uptake for T cell ROS regulation and activation.


Subject(s)
B-Lymphocytes/metabolism , Cystine/metabolism , Lymphocyte Activation , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism , Amino Acid Transport System y+/metabolism , Cell Line, Tumor , Cellular Reprogramming , Flow Cytometry , Fluorescein-5-isothiocyanate/metabolism , Fluorescence , Fluorescent Dyes/metabolism , Glutathione/metabolism , Humans , Microscopy, Fluorescence , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Up-Regulation
6.
Mol Imaging Biol ; 18(1): 18-23, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25971659

ABSTRACT

PURPOSE: Alanine-serine-cysteine transporter 2 (ASCT2) expression has been demonstrated as a promising lung cancer biomarker. (2S,4R)-4-[(18)F]Fluoroglutamine (4-[(18)F]fluoro-Gln) positron emission tomography (PET) was evaluated in preclinical models of non-small cell lung cancer as a quantitative, non-invasive measure of ASCT2 expression. PROCEDURES: In vivo microPET studies of 4-[(18)F]fluoro-Gln uptake were undertaken in human cell line xenograft tumor-bearing mice of varying ASCT2 levels, followed by a genetically engineered mouse model of epidermal growth factor receptor (EGFR)-mutant lung cancer. The relationship between a tracer accumulation and ASCT2 levels in tumors was evaluated by IHC and immunoblotting. RESULT: 4-[(18)F]Fluoro-Gln uptake, but not 2-deoxy-2-[(18)F]fluoro-D-glucose, correlated with relative ASCT2 levels in xenograft tumors. In genetically engineered mice, 4-[(18)F]fluoro-Gln accumulation was significantly elevated in lung tumors, relative to normal lung and cardiac tissues. CONCLUSIONS: 4-[(18)F]Fluoro-Gln PET appears to provide a non-invasive measure of ASCT2 expression. Given the potential of ASCT2 as a lung cancer biomarker, this and other tracers reflecting ASCT2 levels could emerge as precision imaging diagnostics in this setting.


Subject(s)
Amino Acid Transport System ASC/metabolism , Glutamine/analogs & derivatives , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Positron-Emission Tomography/methods , Animals , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/metabolism , ErbB Receptors/genetics , Female , Glutamine/metabolism , Humans , Male , Mice, Nude , Minor Histocompatibility Antigens , Mutation/genetics , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 137(7): 1587-97, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25821004

ABSTRACT

We previously elucidated the pleotropic role of solute carrier family A1 member 5 (SLC1A5) as the primary transporter of glutamine (Gln), a modulator of cell growth and oxidative stress in non-small cell lung cancer (NSCLC). The aim of our study was to evaluate SLC1A5 as a potential new therapeutic target and candidate biomarker predictive of survival and response to therapy. SLC1A5 targeting was examined in a panel of NSCLC and human bronchial cell lines by RNA interference and by a small molecular inhibitor, gamma-l-glutamyl-p-nitroanilide (GPNA). The effects of targeting SLC1A5 on cell growth, Gln uptake, ATP level, autophagy and cell death were examined. Inactivation of SLC1A5 genetically or pharmacologically decreased Gln consumption, inhibited cell growth, induced autophagy and apoptosis in a subgroup of NSCLC cell lines that overexpress SLC1A5. Targeting SLC1A5 function decreased tumor growth in NSCLC xenografts. A multivariate Cox proportional hazards analysis indicates that patients with increased SLC1A5 mRNA expression have significantly shorter overall survival (p = 0.01, HR = 1.24, 95% CI: 1.05-1.46), adjusted for age, gender, smoking history and disease stage. In an immunohistochemistry study on 207 NSCLC patients, SLC1A5 protein expression remained highly significant prognostic value in both univariate (p < 0.0001, HR = 1.45, 95% CI: 1.15-1.50) and multivariate analyses (p = 0.04, HR = 1.22, 95% CI: 1.01-1.31). These results position SLC1A5 as a new candidate prognostic biomarker for selective targeting of Gln-dependent NSCLC.


Subject(s)
Amino Acid Transport System ASC/antagonists & inhibitors , Amino Acid Transport System ASC/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/therapy , Glutamine/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Amino Acid Transport System ASC/biosynthesis , Amino Acid Transport System ASC/genetics , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Minor Histocompatibility Antigens , Molecular Targeted Therapy , Prognosis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
8.
Proc Natl Acad Sci U S A ; 112(11): 3469-74, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25733852

ABSTRACT

Aberrant expression of RNA-binding proteins has profound implications for cellular physiology and the pathogenesis of human diseases such as cancer. We previously identified the Fragile X-Related 1 gene (FXR1) as one amplified candidate driver gene at 3q26-29 in lung squamous cell carcinoma (SCC). FXR1 is an autosomal paralog of Fragile X mental retardation 1 and has not been directly linked to human cancers. Here we demonstrate that FXR1 is a key regulator of tumor progression and its overexpression is critical for nonsmall cell lung cancer (NSCLC) cell growth in vitro and in vivo. We identified the mechanisms by which FXR1 executes its regulatory function by forming a novel complex with two other oncogenes, protein kinase C, iota and epithelial cell transforming 2, located in the same amplicon via distinct binding mechanisms. FXR1 expression is a candidate biomarker predictive of poor survival in multiple solid tumors including NSCLCs. Because FXR1 is overexpressed and associated with poor clinical outcomes in multiple cancers, these results have implications for other solid malignancies.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Lung Neoplasms/genetics , RNA-Binding Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , DNA Copy Number Variations/genetics , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Prognosis , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Survival Analysis , Treatment Outcome
9.
Cancer Prev Res (Phila) ; 7(7): 748-57, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24819876

ABSTRACT

We identified acyl-coenzyme A-binding protein (ACBP) as part of a proteomic signature predicting the risk of having lung cancer. Because ACBP is known to regulate ß-oxidation, which in turn controls cellular proliferation, we hypothesized that ACBP contributes to regulation of cellular proliferation and survival of non-small cell lung cancer (NSCLC) by modulating ß-oxidation. We used matrix-assisted laser desorption/ionization-imaging mass spectrometry (MALDI-IMS) and immunohistochemistry (IHC) to confirm the tissue localization of ABCP in pre-invasive and invasive NSCLCs. We correlated ACBP gene expression levels in NSCLCs with clinical outcomes. In loss-of-function studies, we tested the effect of the downregulation of ACBP on cellular proliferation and apoptosis in normal bronchial and NSCLC cell lines. Using tritiated-palmitate ((3)H-palmitate), we measured ß-oxidation levels and tested the effect of etomoxir, a ß-oxidation inhibitor, on proliferation and apoptosis. MALDI-IMS and IHC analysis confirmed that ACBP is overexpressed in pre-invasive and invasive lung cancers. High ACBP gene expression levels in NSCLCs correlated with worse survival (HR = 1.73). We observed a 40% decrease in ß-oxidation and concordant decreases in proliferation and increases in apoptosis in ACBP-depleted NSCLC cells as compared with bronchial airway epithelial cells. Inhibition of ß-oxidation by etomoxir in ACBP-overexpressing cells produced dose-dependent decrease in proliferation and increase in apoptosis (P = 0.01 and P < 0.001, respectively). These data suggest a role for ACBP in controlling lung cancer progression by regulating ß-oxidation.


Subject(s)
Acetyl Coenzyme A/metabolism , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Diazepam Binding Inhibitor/metabolism , Palmitic Acid/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenosine Triphosphate/metabolism , Blotting, Western , Bronchi/metabolism , Bronchi/pathology , Carcinoma in Situ/metabolism , Carcinoma in Situ/mortality , Carcinoma in Situ/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Humans , Immunoenzyme Techniques , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial , Oxidation-Reduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Clin Cancer Res ; 19(20): 5580-90, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23908357

ABSTRACT

PURPOSE: Chromosome 3q26-29 is a critical region of genomic amplification in lung squamous cell carcinomas (SCC). Identification of candidate drivers in this region could help uncover new mechanisms in the pathogenesis and potentially new targets in SCC of the lung. EXPERIMENTAL DESIGN: We conducted a meta-analysis of seven independent datasets containing a total of 593 human primary SCC samples to identify consensus candidate drivers in 3q26-29 amplicon. Through integrating protein-protein interaction network information, we further filtered for candidates that may function together in a network. Computationally predicted candidates were validated using RNA interference (RNAi) knockdown and cell viability assays. Clinical relevance of the experimentally supported drivers was evaluated in an independent cohort of 52 lung SCC patients using survival analysis. RESULTS: The meta-analysis identified 20 consensus candidates, among which four (SENP2, DCUN1D1, DVL3, and UBXN7) are involved in a small protein-protein interaction network. Knocking down any of the four proteins led to cell growth inhibition of the 3q26-29-amplified SCC. Moreover, knocking down of SENP2 resulted in the most significant cell growth inhibition and downregulation of DCUN1D1 and DVL3. Importantly, a gene expression signature composed of SENP2, DCUN1D1, and DVL3 stratified patients into subgroups with different response to adjuvant chemotherapy. CONCLUSION: Together, our findings show that SENP2, DCUN1D1, and DVL3 are candidate driver genes in the 3q26-29 amplicon of SCC, providing novel insights into the molecular mechanisms of disease progression and may have significant implication in the management of SCC of the lung.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 3 , Gene Amplification , Genomics , Lung Neoplasms/genetics , Oncogenes , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/mortality , Drug Resistance, Neoplasm , Gene Regulatory Networks , Genomics/methods , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Transcriptome
11.
Clin Cancer Res ; 19(3): 560-70, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23213057

ABSTRACT

PURPOSE: We have previously identified solute-linked carrier family A1 member 5 (SLC1A5) as an overexpressed protein in a shotgun proteomic analysis of stage I non-small cell lung cancer (NSCLC) when compared with matched controls. We hypothesized that overexpression of SLC1A5 occurs to meet the metabolic demand for lung cancer cell growth and survival. EXPERIMENTAL DESIGN: To test our hypothesis, we first analyzed the protein expression of SLC1A5 in archival lung cancer tissues by immunohistochemistry and immunoblotting (N = 98) and in cell lines (N = 36). To examine SLC1A5 involvement in amino acid transportation, we conducted kinetic analysis of l-glutamine (Gln) uptake in lung cancer cell lines in the presence and absence of a pharmacologic inhibitor of SLC1A5, gamma-l-Glutamyl-p-Nitroanilide (GPNA). Finally, we examined the effect of Gln deprivation and uptake inhibition on cell growth, cell-cycle progression, and growth signaling pathways of five lung cancer cell lines. RESULTS: Our results show that (i) SLC1A5 protein is expressed in 95% of squamous cell carcinomas (SCC), 74% of adenocarcinomas (ADC), and 50% of neuroendocrine tumors; (ii) SLC1A5 is located at the cytoplasmic membrane and is significantly associated with SCC histology and male gender; (iii) 68% of Gln is transported in a Na(+)-dependent manner, 50% of which is attributed to SLC1A5 activity; and (iv) pharmacologic and genetic targeting of SLC1A5 decreased cell growth and viability in lung cancer cells, an effect mediated in part by mTOR signaling. CONCLUSIONS: These results suggest that SLC1A5 plays a key role in Gln transport controlling lung cancer cells' metabolism, growth, and survival.


Subject(s)
Amino Acid Transport System ASC/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Glutamine/metabolism , Lung Neoplasms/metabolism , Adult , Aged , Amino Acid Transport System ASC/genetics , Biological Transport , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cell Survival/genetics , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Minor Histocompatibility Antigens , RNA Interference , Reactive Oxygen Species/metabolism , Sex Factors , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
12.
BMC Syst Biol ; 7 Suppl 5: S1, 2013.
Article in English | MEDLINE | ID: mdl-24564859

ABSTRACT

BACKGROUND: Oncogenic mechanisms in small-cell lung cancer remain poorly understood leaving this tumor with the worst prognosis among all lung cancers. Unlike other cancer types, sequencing genomic approaches have been of limited success in small-cell lung cancer, i.e., no mutated oncogenes with potential driver characteristics have emerged, as it is the case for activating mutations of epidermal growth factor receptor in non-small-cell lung cancer. Differential gene expression analysis has also produced SCLC signatures with limited application, since they are generally not robust across datasets. Nonetheless, additional genomic approaches are warranted, due to the increasing availability of suitable small-cell lung cancer datasets. Gene co-expression network approaches are a recent and promising avenue, since they have been successful in identifying gene modules that drive phenotypic traits in several biological systems, including other cancer types. RESULTS: We derived an SCLC-specific classifier from weighted gene co-expression network analysis (WGCNA) of a lung cancer dataset. The classifier, termed SCLC-specific hub network (SSHN), robustly separates SCLC from other lung cancer types across multiple datasets and multiple platforms, including RNA-seq and shotgun proteomics. The classifier was also conserved in SCLC cell lines. SSHN is enriched for co-expressed signaling network hubs strongly associated with the SCLC phenotype. Twenty of these hubs are actionable kinases with oncogenic potential, among which spleen tyrosine kinase (SYK) exhibits one of the highest overall statistical associations to SCLC. In patient tissue microarrays and cell lines, SCLC can be separated into SYK-positive and -negative. SYK siRNA decreases proliferation rate and increases cell death of SYK-positive SCLC cell lines, suggesting a role for SYK as an oncogenic driver in a subset of SCLC. CONCLUSIONS: SCLC treatment has thus far been limited to chemotherapy and radiation. Our WGCNA analysis identifies SYK both as a candidate biomarker to stratify SCLC patients and as a potential therapeutic target. In summary, WGCNA represents an alternative strategy to large scale sequencing for the identification of potential oncogenic drivers, based on a systems view of signaling networks. This strategy is especially useful in cancer types where no actionable mutations have emerged.


Subject(s)
Gene Expression Profiling , Gene Regulatory Networks , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncogene Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Molecular Targeted Therapy , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Proteomics , Syk Kinase
13.
Mol Cell Proteomics ; 11(10): 916-32, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22761400

ABSTRACT

Advances in proteomic analysis of human samples are driving critical aspects of biomarker discovery and the identification of molecular pathways involved in disease etiology. Toward that end, in this report we are the first to use a standardized shotgun proteomic analysis method for in-depth tissue protein profiling of the two major subtypes of nonsmall cell lung cancer and normal lung tissues. We identified 3621 proteins from the analysis of pooled human samples of squamous cell carcinoma, adenocarcinoma, and control specimens. In addition to proteins previously shown to be implicated in lung cancer, we have identified new pathways and multiple new differentially expressed proteins of potential interest as therapeutic targets or diagnostic biomarkers, including some that were not identified by transcriptome profiling. Up-regulation of these proteins was confirmed by multiple reaction monitoring mass spectrometry. A subset of these proteins was found to be detectable and differentially present in the peripheral blood of cases and matched controls. Label-free shotgun proteomic analysis allows definition of lung tumor proteomes, identification of biomarker candidates, and potential targets for therapy.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Chromatography, Liquid , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Mass Spectrometry , Neoplasm Proteins/metabolism , Neoplasm Staging , Proteomics/methods , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL