Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 466
Filter
1.
Oncogene ; 33(36): 4464-73, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-24096489

ABSTRACT

In an effort to identify novel biallelically inactivated tumor suppressor genes (TSGs) in sporadic invasive and preinvasive non-small-cell lung cancer (NSCLC) genomes, we applied a comprehensive integrated multiple 'omics' approach to investigate patient-matched, paired NSCLC tumor and non-malignant parenchymal tissues. By surveying lung tumor genomes for genes concomitantly inactivated within individual tumors by multiple mechanisms, and by the frequency of disruption in tumors across multiple cohorts, we have identified a putative lung cancer TSG, Eyes Absent 4 (EYA4). EYA4 is frequently and concomitantly deleted, hypermethylated and underexpressed in multiple independent lung tumor data sets, in both major NSCLC subtypes and in the earliest stages of lung cancer. We found that decreased EYA4 expression is not only associated with poor survival in sporadic lung cancers but also that EYA4 single-nucleotide polymorphisms are associated with increased familial cancer risk, consistent with EYA4s proximity to the previously reported lung cancer susceptibility locus on 6q. Functionally, we found that EYA4 displays TSG-like properties with a role in modulating apoptosis and DNA repair. Cross-examination of EYA4 expression across multiple tumor types suggests a cell-type-specific tumorigenic role for EYA4, consistent with a tumor suppressor function in cancers of epithelial origin. This work shows a clear role for EYA4 as a putative TSG in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/epidemiology , Gene Silencing , Lung Neoplasms/pathology , Trans-Activators/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Chromosomes, Human, Pair 6 , DNA Methylation , Epigenesis, Genetic , Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Frequency , Genes, Tumor Suppressor , Genetic Association Studies , Genetic Variation , Genome, Human , Humans , Lung Neoplasms/genetics , Polymorphism, Single Nucleotide , Trans-Activators/metabolism , Tumor Cells, Cultured
2.
Oncogene ; 32(34): 4034-42, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-22964644

ABSTRACT

KRAS mutations are one of the most common driver mutations in non-small-cell lung cancer (NSCLC) and finding druggable target molecules to inhibit oncogenic KRAS signaling is a significant challenge in NSCLC therapy. We recently identified epiregulin (EREG) as one of several putative transcriptional targets of oncogenic KRAS signaling in both KRAS-mutant NSCLC cells and immortalized bronchial epithelial cells expressing ectopic mutant KRAS. In the current study, we found that EREG is overexpressed in NSCLCs harboring KRAS, BRAF or EGFR mutations compared with NSCLCs with wild-type KRAS/BRAF/EGFR. Small interfering RNAs (siRNAs) targeting mutant KRAS, but not an siRNA targeting wild-type KRAS, significantly reduced EREG expression in KRAS-mutant and EREG-overexpressing NSCLC cell lines. In these cell lines, EREG expression was downregulated by MEK and ERK inhibitors. Importantly, EREG expression significantly correlated with KRAS expression or KRAS copy number in KRAS-mutant NSCLC cell lines. Further expression analysis using 89 NSCLC specimens showed that EREG was predominantly expressed in NSCLCs with pleural involvement, lymphatic permeation or vascular invasion and in KRAS-mutant adenocarcinomas. In addition, multivariate analysis revealed that EREG expression is an independent prognostic marker and EREG overexpression in combination with KRAS mutations was associated with an unfavorable prognosis for lung adenocarcinoma patients. In KRAS-mutant and EREG overexpressing NSCLC cells, siRNA-mediated EREG silencing inhibited anchorage-dependent and -independent growth and induced apoptosis. Our findings suggest that oncogenic KRAS-induced EREG overexpression contributes to an aggressive phenotype and could be a promising therapeutic target in oncogenic KRAS-driven NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epidermal Growth Factor/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Aged , Apoptosis/genetics , Butadienes/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Epiregulin , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Mutation , Nitriles/pharmacology , Phenotype , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Pyrazoles/pharmacology , Pyridazines/pharmacology , RNA Interference , ras Proteins/metabolism
3.
Ann Oncol ; 21 Suppl 7: vii225-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20943619

ABSTRACT

Until recently the major clinical question was 'Is it small-cell or non small-cell cancer'. However, advances in conventional and targeted therapy have completely changed the landscape. Identification of the major non-small-cell lung cancer (NSCLC) types (adenocarcinoma and squamous carcinoma) are important for a number of predictive and prognostic reasons, including pemetrexed treatment, anti-angiogenic therapy and administration of tyrosine kinase inhibitors. Fortunately, advances in pathology of lung cancer have kept abreast, with newer, simplified methods to identify the major NSCLC types in small diagnostic samples, and modifications of the pathological classification of adenocarcinomas reflecting changing clinical and molecular concepts. For the patient to obtain maximum benefit from the recent developments in therapeutics, a multidisciplinary approach is required with co-operation between oncologists, surgeons, radiologists and pathologists.


Subject(s)
Carcinoma, Non-Small-Cell Lung/classification , Lung Neoplasms/classification , Terminology as Topic , Adenocarcinoma/diagnosis , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/diagnosis , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Molecular Diagnostic Techniques , Mutation/physiology
4.
Curr Mol Med ; 10(2): 123-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20196733

ABSTRACT

DNA methylation plays a critical role in the regulation of gene expression, differentiation and in the development of cancer and other diseases. Hypermethylation of CpG islands located in the promoter regions of tumor suppressor genes is now firmly established as the most frequent mechanism for gene inactivation in cancers. Feasibility of using DNA methylation based biomarkers for early detection of cancer has been shown. Potential of using DNA methylation for prediction of therapeutic outcome and patient survival has also been shown. DNA originated from cancer cells has been routinely detected in clinical specimens (ex. Plasma/serum, sputum, urine etc.) from cancer patients. Presence of methylated DNA sequences in clinical specimens and potential of using them as biomarkers have been recognized. Novel methylation based biomarkers that can be used in clinical specimens, obtained non-invasively from cancer patients, offer significant practical advantages. More resources need to be committed to this area of biomarker research. Thus, we review recent findings on DNA methylation based cancer biomarkers with particular focus on these applicable to the clinical specimens obtained non-invasively from cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , DNA Methylation , Early Detection of Cancer/methods , Medical Oncology/methods , Neoplasms/blood , Neoplasms/diagnosis , Neoplasms/genetics , Algorithms , CpG Islands , Female , Humans , Male , Neoplasms/metabolism , Promoter Regions, Genetic , Treatment Outcome
5.
Oncogene ; 29(18): 2616-27, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20154724

ABSTRACT

Recent studies have established that amplification of the MET proto-oncogene can cause resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC) cell lines with EGFR-activating mutations. The role of non-amplified MET in EGFR-dependent signaling before TKI resistance, however, is not well understood. Using NSCLC cell lines and transgenic models, we demonstrate here that EGFR activation by either mutation or ligand binding increases MET gene expression and protein levels. Our analysis of 202 NSCLC patient specimens was consistent with these observations: levels of MET were significantly higher in NSCLC with EGFR mutations than in NSCLC with wild-type EGFR. EGFR regulation of MET levels in cell lines occurred through the hypoxia-inducible factor (HIF)-1alpha pathway in a hypoxia-independent manner. This regulation was lost, however, after MET gene amplification or overexpression of a constitutively active form of HIF-1alpha. EGFR- and hypoxia-induced invasiveness of NSCLC cells, but not cell survival, were found to be MET dependent. These findings establish that, absent MET amplification, EGFR signaling can regulate MET levels through HIF-1alpha and that MET is a key downstream mediator of EGFR-induced invasiveness in EGFR-dependent NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-met/physiology , Receptors, Growth Factor/physiology , Animals , Cell Hypoxia , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Gene Amplification , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Mice , Neoplasm Invasiveness , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/analysis , Proto-Oncogene Proteins c-met/genetics , Receptors, Growth Factor/analysis , Receptors, Growth Factor/genetics
6.
Oncogene ; 29(10): 1421-30, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-19966867

ABSTRACT

DNA amplifications, leading to the overexpression of oncogenes, are a cardinal feature of lung cancer and directly contribute to its pathogenesis. To uncover such novel alterations, we performed an array-based comparative genomic hybridization survey of 128 non-small-cell lung cancer cell lines and tumors. Prominent among our findings, we identified recurrent high-level amplification at cytoband 22q11.21 in 3% of lung cancer specimens, with another 11% of specimens exhibiting low-level gain spanning that locus. The 22q11.21 amplicon core contained eight named genes, only four of which were overexpressed (by transcript profiling) when amplified. Among these, CRKL encodes an adapter protein functioning in signal transduction, best known as a substrate of the BCR-ABL kinase in chronic myelogenous leukemia. RNA-interference-mediated knockdown of CRKL in lung cancer cell lines with (but not without) amplification led to significantly decreased cell proliferation, cell-cycle progression, cell survival, and cell motility and invasion. In addition, overexpression of CRKL in immortalized human bronchial epithelial cells led to enhanced growth factor-independent cell growth. Our findings indicate that amplification and resultant overexpression of CRKL contribute to diverse oncogenic phenotypes in lung cancer, with implications for targeted therapy, and highlight a role of adapter proteins as primary genetic drivers of tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Amplification , Gene Expression Profiling , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Chromosomes, Human, Pair 22/genetics , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Oligonucleotide Array Sequence Analysis , RNA Interference
7.
Oncogene ; 28 Suppl 1: S24-31, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19680293

ABSTRACT

The epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs), gefitinib and erlotinib, are reversible competitive inhibitors of the tyrosine kinase domain of EGFR that bind to its adenosine-5' triphosphate-binding site. Somatic activating mutations of the EGFR gene, increased gene copy number and certain clinical and pathological features have been associated with dramatic tumor responses and favorable clinical outcomes with these agents in patients with non-small-cell lung cancer (NSCLC). The specific types of activating mutations that confer sensitivity to EGFR TKIs are present in the tyrosine kinase (TK) domain of the EGFR gene. Exon 19 deletion mutations and the single-point substitution mutation L858R in exon 21 are the most frequent in NSCLC and are termed 'classical' mutations. The NSCLC tumors insensitive to EGFR TKIs include those driven by the KRAS and MET oncogenes. Most patients who initially respond to gefitinib and erlotinib eventually become resistant and experience progressive disease. The point mutation T790M accounts for about one half of these cases of acquired resistance. Various second-generation EGFR TKIs are currently being evaluated and may have the potential to overcome T790M-mediated resistance by virtue of their irreversible inhibition of the receptor TK domain.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gefitinib , Gene Amplification , Humans , Lung Neoplasms/genetics , Protein Structure, Tertiary/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met , Proto-Oncogene Proteins p21(ras) , Quinazolines/therapeutic use , Receptors, Growth Factor/genetics , Sequence Deletion , Treatment Outcome , ras Proteins/genetics
9.
Oncogene ; 27(33): 4615-24, 2008 Jul 31.
Article in English | MEDLINE | ID: mdl-18391978

ABSTRACT

Chromosomal translocation is the best-characterized genetic mechanism for oncogene activation. However, there are documented examples of activation by alternate mechanisms, for example gene dosage increase, though its prevalence is unclear. Here, we answered the fundamental question of the contribution of DNA amplification as a molecular mechanism driving oncogenesis. Comparing 104 cancer lines representing diverse tissue origins identified genes residing in amplification 'hotspots' and discovered an unexpected frequency of genes activated by this mechanism. The 3431 amplicons identified represent approximately 10 per hematological and approximately 36 per epithelial cancer genome. Many recurrently amplified oncogenes were previously known to be activated only by disease-specific translocations. The 135 hotspots identified contain 538 unique genes and are enriched for proliferation, apoptosis and linage-dependency genes, reflecting functions advantageous to tumor growth. Integrating gene dosage with expression data validated the downstream impact of the novel amplification events in both cell lines and clinical samples. For example, multiple downstream components of the EGFR-family-signaling pathway, including CDK5, AKT1 and SHC1, are overexpressed as a direct result of gene amplification in lung cancer. Our findings suggest that amplification is far more common a mechanism of oncogene activation than previously believed and that specific regions of the genome are hotspots of amplification.


Subject(s)
Gene Amplification/genetics , Gene Dosage/genetics , Lung Neoplasms/genetics , Oncogene Proteins/genetics , Oncogenes/genetics , Translocation, Genetic/genetics , Animals , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Genome, Human , Humans , Lung Neoplasms/metabolism , Oncogene Proteins/metabolism , Signal Transduction/genetics
10.
Oncogene ; 27(25): 3635-40, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18212743

ABSTRACT

Lung cancer is a leading cause of cancer death, where the amplification of oncogenes contributes to tumorigenesis. Genomic profiling of 128 lung cancer cell lines and tumors revealed frequent focal DNA amplification at cytoband 14q13.3, a locus not amplified in other tumor types. The smallest region of recurrent amplification spanned the homeobox transcription factor TITF1 (thyroid transcription factor 1; also called NKX2-1), previously linked to normal lung development and function. When amplified, TITF1 exhibited increased expression at both the RNA and protein levels. Small interfering RNA (siRNA)-mediated knockdown of TITF1 in lung cancer cell lines with amplification led to reduced cell proliferation, manifested by both decreased cell-cycle progression and increased apoptosis. Our findings indicate that TITF1 amplification and overexpression contribute to lung cancer cell proliferation rates and survival and implicate TITF1 as a lineage-specific oncogene in lung cancer.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/biosynthesis , Transcription Factors/biosynthesis , Apoptosis , Cell Line, Tumor , Cell Lineage , Chromosome Mapping , Genome, Human , Humans , Models, Biological , Neoplasm Metastasis , Oligonucleotide Array Sequence Analysis , Oncogenes , RNA, Small Interfering/metabolism , Thyroid Nuclear Factor 1
11.
Ann Oncol ; 18(4): 752-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17317677

ABSTRACT

BACKGROUND: Biological markers for optimal selection of patient to epidermal growth factor receptor (EGFR)-targeted therapies are not established in advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: EGFR/HER2 gene copy number by FISH, EGFR protein and pAKT expression by immunohistochemistry (IHC) and EGFR and KRAS mutations were tested in 204 gefitinib-treated NSCLC patients. RESULTS: Increased EGFR and HER2 gene copy number (FISH+), EGFR protein overexpression (IHC+), EGFR mutations and pAKT overexpression were all associated with significantly higher response rates (33%, 29%, 22%, 39% and 20% respectively). EGFR FISH+ (32%) and IHC+ (61%) correlated with improved survival, while EGFR mutations (27%), KRAS mutations (26%) and pAKT expression (69%) did not. In multivariate survival analysis EGFR FISH and IHC were independent predictive markers. EGFR FISH+/IHC+ patients (23%) had a median survival of 21 months versus 6 months for double-negative patients (30%). CONCLUSION: Combination of EGFR FISH and IHC is effective predictor for benefit from gefitinib. Patients with double-negative results are unlikely to benefit in western NSCLC populations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Gene Dosage , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , ErbB Receptors/analysis , Female , Gefitinib , Genes, erbB-2 , Genes, ras , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Mutation , Treatment Outcome
12.
Br J Cancer ; 94(12): 1927-35, 2006 Jun 19.
Article in English | MEDLINE | ID: mdl-16705311

ABSTRACT

Lung cancer is the leading cause of cancer-related mortality in the world, with small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) comprising the two major cell types. Although these cell types can be distinguished readily at the histological level, knowledge of their underlying molecular differences is very limited. In this study, we compared 14 SCLC cell lines against 27 NSCLC cell lines using an integrated array comparative genomic hybridisation and gene expression profiling approach to identify subtype-specific disruptions. Using stringent criteria, we have identified 159 of the genes that are responsible for the different biology of these cell types. Sorting of these genes by their biological functions revealed the differential disruption of key components involved in cell cycle pathways. Our novel comparative combined genome and transcriptome analysis not only identified differentially altered genes, but also revealed that certain shared pathways are preferentially disrupted at different steps in these cell types. Small cell lung cancer exhibited increased expression of MRP5, activation of Wnt pathway inhibitors, and upregulation of p38 MAPK activating genes, while NSCLC showed downregulation of CDKN2A, and upregulation of MAPK9 and EGFR. This information suggests that cell cycle upregulation in SCLC and NSCLC occurs through drastically different mechanisms, highlighting the need for differential molecular target selection in the treatment of these cancers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Small Cell/genetics , Cell Cycle/physiology , Genes, Neoplasm , Lung Neoplasms/genetics , Cell Line, Tumor , Gene Dosage , Gene Expression , Gene Expression Profiling , Humans , Nucleic Acid Hybridization , Principal Component Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
Br J Cancer ; 93(9): 1029-37, 2005 Oct 31.
Article in English | MEDLINE | ID: mdl-16234815

ABSTRACT

The transforming growth factor beta (TGFbeta)-signalling pathway is deregulated in many cancers. We examined the role of gene silencing via aberrant methylation of DRM/Gremlin and HPP1, which inhibit TGFbeta signalling, and RUNX3, which facilitates TGFbeta-signalling, of all genes that are thought to be tumour suppressors, are aberrantly expressed, and are thus thought to have important role in human cancers. We examined DRM/Gremlin mRNA expression in 44 cell lines and the promoter methylation status of DRM/Gremlin, HPP1, and RUNX3 in 44 cell lines and 511 primary tumours. The loss of DRM/Gremlin mRNA expression in human cancer cell lines is associated with DNA methylation, and treatment with the methylation inhibitor-reactivated mRNA expression (n=13). Methylation percentages of the three genes ranged from 0-83% in adult tumours and 0-50% in paediatric tumours. Methylation of DRM/Gremlin was more frequent in lung tumours in smokers, and methylation of all three genes was inversely correlated with prognosis in patients with bladder or prostate cancer. Our results provide strong evidence that these TGFbeta-related genes are frequently deregulated through aberrant methylation in many human malignancies.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Aged , Core Binding Factor Alpha 3 Subunit/metabolism , Down-Regulation , Female , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Tumor Cells, Cultured
14.
J Med Genet ; 42(7): 558-64, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15994877

ABSTRACT

BACKGROUND: Somatically acquired chromosomal translocation is a common mechanism of oncogene activation in many haematopoietic tumours and sarcomas. However, very few recurrent chromosomal translocations have been reported in more common epithelial tumours such as lung carcinomas. METHODS: We established a cell line HCC2429 from an aggressive, metastatic lung cancer arising in a young, non-smoking woman, demonstrating a t(15;19)(q13.2;p13.1). The breakpoints on chromosomes 15 and 19 were cloned using long distance inverse PCR and we determined by RT-PCR that the translocation results in a novel fusion transcript in which the 3' end Brd4 on chromosome 19p is fused to the 5' end of NUT on chromosome 15q. RESULTS: In total, 128 lung cancer tissues were screened using fluorescent in situ hybridisation, but none of the tumours screened demonstrated t(15;19), suggesting that this translocation is not commonly found in lung cancer. Consistent with previous literature, ectopic expression of wild type Brd4 was shown to inhibit G(1) to S progression. However, we also found that the Brd4-NUT fusion augments the inhibition of progression to S phase compared with wild type Brd4. CONCLUSION: Alteration in cell cycle kinetics is important in tumorigenesis, although the exact role of Brd4-NUT fusion protein in the pathogenesis of lung cancers remains unclear and need to be further elucidated.


Subject(s)
Lung Neoplasms/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , S Phase/genetics , Translocation, Genetic/genetics , Adult , Blotting, Northern , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Mutational Analysis , Female , Humans , Nuclear Proteins/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Polymerase Chain Reaction , Transfection
15.
Br J Cancer ; 92(5): 942-8, 2005 Mar 14.
Article in English | MEDLINE | ID: mdl-15756262

ABSTRACT

SPARC (secreted protein acidic and rich in cysteine) is an extracellular Ca2+-binding matricellular glycoprotein associated with the regulation of cell adhesion and growth. We investigated loss of expression of SPARC gene and promoter methylation in lung cancers and correlated the data with clinicopathological features. We observed loss of SPARC expression in 12 of 20 (60%) lung cancer cell lines. Treatment of expression-negative cell lines with a demethylating agent restored expression in all cases. Methylation frequencies of SPARC gene were 55% in 20 lung cancer cell lines. Primary tumours had methylation at a rate of 69% (119 of 173), while nonmalignant lung tissues (n=60) had very low rates (3%). In lung adenocarcinomas, SPARC methylation correlated with a negative prognosis (P=0.0021; relative risk 4.65, 95% confidence interval 1.75-12.35, multivariate Cox's proportional-hazard model). Immunostaining revealed protein expression in bronchial epithelium (weak intensity) and in juxtatumoral stromal tissues (strong intensity) accompanied by frequent loss in cancer cells that correlated with the presence of methylation (P<0.001). Our findings are of biological interest and potentially of clinical importance in human lung cancers.


Subject(s)
DNA Methylation , DNA, Neoplasm/genetics , Lung Neoplasms/genetics , Osteonectin/genetics , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Transcription, Genetic
16.
Br J Cancer ; 92(8): 1553-60, 2005 Apr 25.
Article in English | MEDLINE | ID: mdl-15785753

ABSTRACT

Genetic alterations occurring on human chromosome arm 1p are common in many types of cancer including lung, breast, neuroblastoma, pheochromocytoma, and colorectal. The identification of tumour suppressors and oncogenes on this arm has been limited by the low resolution of current technologies for fine mapping. In order to identify genetic alterations on 1p in small-cell lung carcinoma, we developed a new resource for fine mapping segmental DNA copy number alterations. We have constructed an array of 642 ordered and fingerprint-verified bacterial artificial chromosome clones spanning the 120 megabase (Mb) 1p arm from 1p11.2 to p36.33. The 1p arm of 15 small-cell lung cancer cell lines was analysed at sub-Mb resolution using this arm-specific array. Among the genetic alterations identified, two regions of recurrent amplification emerged. They were detected in at least 45% of the samples: a 580 kb region at 1p34.2-p34.3 and a 270 kb region at 1p11.2. We further defined the potential importance of these genomic amplifications by analysing the RNA expression of the genes in these regions with Affymetrix oligonucleotide arrays and semiquantitative reverse transcriptase-polymerase chain reaction. Our data revealed overexpression of the genes HEYL, HPCAL4, BMP8, IPT, and RLF, coinciding with genomic amplification.


Subject(s)
Carcinoma, Small Cell/genetics , Chromosomes, Human, Pair 1/genetics , Gene Expression Profiling , Lung Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Gene Amplification , Humans , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
17.
Br J Cancer ; 92(6): 1117-25, 2005 Mar 28.
Article in English | MEDLINE | ID: mdl-15770214

ABSTRACT

The human DOC-2/DAB2 interactive protein (hDAB2IP) gene is a novel member of the Ras GTPase-activating family and has been demonstrated to be a tumour-suppressor gene inactivated by methylation in several cancers. In this study, we analysed the methylation and expression status of hDAB2IP in gastrointestinal tumours. The promoter region of hDAB2IP was divided into two regions (m2a and m2b) based on our previous report, and the methylation status was determined by bisulphite DNA sequencing in gastric cancer cell lines. The gene expression was semiquantified by real-time RT-PCR, and the results indicated that the m2b promoter region might be an authentic methylation-mediated key regulator of the gene expression. Based on the sequence data, we developed a methylation-specific PCR (MSP) for the m2a and m2b regions and applied it to the samples. Methylation-specific PCR revealed aberrant methylation in the m2a region in eight of 12 gastric cancer cell lines (67%), 16 of 35 gastric cancer tissues (46%) and 29 of 60 colorectal cancer tissues (48%), and in the m2b region in eight of 12 cell lines (67%), 15 of 35 gastric cancer tissues (43%) and 28 of 60 colorectal cancer tissues (47%). On the other hand, seven (12%) and 11 (19%) of 59 gastrointestinal nonmalignant mucosal specimens showed methylation in the m2a and m2b regions, respectively, suggesting that hDAB2IP methylation might play a causative role in carcinogenesis. The 5-aza-2'-deoxycytidine treatment restored the gene expression in the m2b-methylated cell lines, confirming that the methylation caused gene downregulation. We also examined the relationship between hDAB2IP methylation and the clinicopathological features in patients with primary tumours, and determined that methylation in the m2b region was associated with location of the tumour in the stomach. In summary, our results demonstrated that hDAB2IP methylation is frequently present in gastrointestinal tumours and that the resulting gene silencing plays an important role in gastrointestinal carcinogenesis.


Subject(s)
Azacitidine/analogs & derivatives , DNA Methylation , Gastrointestinal Neoplasms/genetics , Promoter Regions, Genetic , ras GTPase-Activating Proteins/genetics , Azacitidine/pharmacology , Cell Line, Tumor , Decitabine , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Humans , Hydroxamic Acids/pharmacology , Sequence Analysis, DNA
18.
Am J Hum Genet ; 75(3): 460-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15272417

ABSTRACT

Lung cancer is a major cause of death in the United States and other countries. The risk of lung cancer is greatly increased by cigarette smoking and by certain occupational exposures, but familial factors also clearly play a major role. To identify susceptibility genes for familial lung cancer, we conducted a genomewide linkage analysis of 52 extended pedigrees ascertained through probands with lung cancer who had several first-degree relatives with the same disease. Multipoint linkage analysis, under a simple autosomal dominant model, of all 52 families with three or more individuals affected by lung, throat, or laryngeal cancer, yielded a maximum heterogeneity LOD score (HLOD) of 2.79 at 155 cM on chromosome 6q (marker D6S2436). A subset of 38 pedigrees with four or more affected individuals yielded a multipoint HLOD of 3.47 at 155 cM. Analysis of a further subset of 23 multigenerational pedigrees with five or more affected individuals yielded a multipoint HLOD score of 4.26 at the same position. The 14 families with only three affected relatives yielded negative LOD scores in this region. A predivided samples test for heterogeneity comparing the LOD scores from the 23 multigenerational families with those from the remaining families was significant (P=.007). The 1-HLOD multipoint support interval from the multigenerational families extends from C6S1848 at 146 cM to 164 cM near D6S1035, overlapping a genomic region that is deleted in sporadic lung cancers as well as numerous other cancer types. Parametric linkage and variance-components analysis that incorporated effects of age and personal smoking also supported linkage in this region, but with somewhat diminished support. These results localize a major susceptibility locus influencing lung cancer risk to 6q23-25.


Subject(s)
Chromosomes, Human, Pair 6 , Genetic Predisposition to Disease , Lung Neoplasms/genetics , Chromosome Mapping , Family Health , Genetic Linkage , Genetic Markers , Genome, Human , Genotype , Humans , Lod Score
19.
Br J Cancer ; 91(4): 771-4, 2004 Aug 16.
Article in English | MEDLINE | ID: mdl-15266335

ABSTRACT

The present study examined the relationship between methylation of five genes (p16(INK4a), RASSF1A, APC, RARbeta and CDH13) and patient survival in 351 cases of surgically resected lung cancers. While there was no relationship between the other genes and survival, p16(INK4a) methylation was significantly related to unfavourable prognosis in lung adenocarcinomas.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA Methylation , DNA, Neoplasm/metabolism , Genes, p16 , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies , Survival Analysis
20.
Thorax ; 58(10): 892-900, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14514947

ABSTRACT

It has been hypothesised that clinically evident lung cancers have accumulated many different genetic or epigenetic abnormalities in oncogenes and/or tumour suppressor genes. This notion has important clinical ramifications. Recent developments in our knowledge of the molecular biology of lung cancer are reviewed, with particular reference to genetic abnormalities in tumour suppressor gene inactivation and overactivity of growth promoting oncogenes. These changes lead to the "hallmarks of lung cancer". These hallmarks are the new rational targets for early detection, prevention, and treatment of lung cancer.


Subject(s)
Lung Neoplasms/genetics , Apoptosis/genetics , DNA Methylation , Genes, Tumor Suppressor , Growth Substances/genetics , Humans , Molecular Biology , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neovascularization, Pathologic/genetics , Proto-Oncogenes/genetics , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...