Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Anim Cells Syst (Seoul) ; 27(1): 72-82, 2023.
Article in English | MEDLINE | ID: mdl-37033451

ABSTRACT

Airborne particulate matter (PM) is a major health hazard worldwide and is a key factor in lung cancer, which remains the most common type of malignancy and the leading cause of cancer-related deaths. DNA methylation is a critical mechanism underlying the detrimental effects of PM, however, the molecular link between PM exposure and lung cancer remains to be elucidated. N-α-acetyltransferase 10 (NAA10) is involved in the cell cycle, migration, apoptosis, differentiation, and proliferation. In order to investigate the role of NAA10 in PM-induced pathogenesis processes leading to lung cancer, we determined the expression and methylation of NAA10 in normal human bronchial epithelial (NHBE) cells treated with PM10, PM10-polycyclic aromatic hydrocarbons (PAH), and PM2.5 and evaluated the prognostic value of the NAA10 methylation status in lung cancer patients. Exposure to all PM types significantly increased the expression of NAA10 mRNA and decreased the methylation of the NAA10 promoter in NHBE cells compared with the mock-treated control. NAA10 hypomethylation was observed in 9.3% (13/140) of lung cancer tissue samples and correlated with NAA10 transcriptional upregulation. Univariate and multivariate analyses revealed that NAA10 hypomethylation was associated with decreased survival of patients with lung cancer. Therefore, these results suggest that PM-induced hypomethylation of the NAA10 may play an important role in the pathogenesis of lung cancer and may be used as a potential prognostic biomarker for lung cancer progression. Further studies with large numbers of patients are warranted to confirm our findings.

2.
Lung Cancer ; 86(1): 85-90, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25172267

ABSTRACT

OBJECTIVES: Lung cancer is the leading cause of cancer-related deaths and is currently a major health problem owing to difficulties in diagnosis at the early stage of the disease. Changes in DNA methylation status have now been identified as a critical component in the initiation of lung cancer, and the detection of DNA methylation is expected to be an important method for the early diagnosis of lung cancer. Nicotine, the principal tobacco alkaloid, directly contributes to lung carcinogenesis through the activation of nicotinic acetylcholine receptors (nAchRs). MATERIALS AND METHODS: To investigate the role of the CHRNB4 gene, which encodes the nAchR ß4 subunit that is ubiquitously expressed on lung epithelial cells, we analyzed its methylation status in 266 patients with non-small cell lung cancer (NSCLC) by using methylation-specific polymerase chain reaction and compared it with clinicopathological parameters. RESULTS AND CONCLUSION: The frequency of CHRNB4 unmethylation was 13.5% and 8.3% in malignant and nonmalignant tissues, respectively. CHRNB4 demethylation was associated with upregulation of its mRNA expression and was more frequent in squamous cell carcinoma and pathological stages II-IIIA disease than in adenocarcinoma and pathological stage I disease, respectively (P=0.003 and P=0.01, respectively). Univariate and multivariate analyses showed that CHRNB4 unmethylation was significantly associated with unfavorable overall survival in the entire patient group as well as in men and ever-smokers. These results suggest that epigenetic regulation of CHRNB4 may affect tumor progression and survival in patients with NSCLC. Further investigation into the molecular basis of the role of CHRNB4 in the progression of NSCLC is warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , DNA Methylation , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Nerve Tissue Proteins/genetics , Receptors, Nicotinic/genetics , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , CpG Islands , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Patient Outcome Assessment , Polymorphism, Single Nucleotide , Prognosis , Risk Factors
3.
Mol Cells ; 36(1): 69-73, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23686431

ABSTRACT

Lung cancer is a leading cause of cancer-related mortality across the world and tobacco smoking is the major risk factor. The Wnt signaling pathway is known to be involved in smoke-induced tumorigenesis in the lung. Promoter hypermethylation of Wnt inhibitory factor 1 (Wif1) has become a common event in a number of human tumors. Using a methylation-specific PCR, hypermethylation of the Wif1 gene promoter was evaluated in 139 primary nonsmall cell lung cancers (NSCLCs) and its correlation with clinicopathological and prognostic parameters was evaluated. Methylation of Wif1 was observed in 47.5% and 20.9% of neoplastic and adjacent normal lung tissues, respectively. Its methylation rate tended to be higher in stage I than stages II-IIIA. Results of Kaplan-Meier analysis showed no significant difference in overall survival according to Wif1 methylation status. However, Wif1 methylation showed an association with unfavorable prognosis of adenocarcinoma (AC) patients with EGFR mutation. According to our current findings, Wif1 promoter methylation is an early, frequent event as an epigenetic field manner and could be considered as a useful prognostic marker for AC patients with EGFR mutation. Further investigation into the therapeutic potential of this finding is warranted.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Mutation/genetics , Repressor Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism
4.
Lung Cancer ; 81(2): 194-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23664447

ABSTRACT

INTRODUCTION: Liver kinase 1 (LKB1) plays a critical barrier role in lung tumorigenesis by controlling initiation, differentiation and metastasis. We searched for genetic and epigenetic alterations of the LKB1 gene in Korean non-small cell lung cancers (NSCLCs) and correlated the results with clinicopathological features. We also investigated the relationship between genetic and epigenetic alterations of LKB1 and mutations in the TP53 gene and epidermal growth factor receptor (EGFR) pathway genes. METHODS: A total of 159 NSCLCs were analyzed for loss of heterozygosity (LOH) at microsatellite loci D19S886, and D19S878. Mutations and methylation status of LKB1 were examined by direct sequencing and a methylation-specific polymerase chain reaction, respectively. RESULTS: A somatic mutation was found in one of the 159 tumors. LOH and promoter methylation was detected in 19.5% (31/159) and 13.2% (21/159) of the tumors, respectively. Four of the 159 tumors had concomitant LOH and methylation of LKB1. In total, 30.2% of the 159 NSCLCs harbored LKB1 LOH or promoter methylation, which were correlated with down-regulation of gene expression. LKB1 LOH was more frequent in males, smokers, and tumors with a TP53 mutation than in females, never-smokers, and tumors without a TP53 mutation, respectively. However, no significant correlation between LKB1 alterations and mutations in EGFR pathway genes was found. CONCLUSION: These results suggest that the prevalence of LKB1 genetic and epigenetic alterations in NSCLCs vary depending on patient ethnicity. Our results show that LKB1 alterations often occur simultaneously with mutations in EGFR pathway genes.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Genes, p53 , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Asian People/genetics , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , DNA Methylation , Down-Regulation , Epigenesis, Genetic , Female , Gene Expression , Humans , Lung Neoplasms/enzymology , Male , Middle Aged , Mutation , Promoter Regions, Genetic , Republic of Korea , Signal Transduction
5.
Mol Cells ; 34(2): 171-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22814847

ABSTRACT

Lung cancer remains a global health problem with a high mortality rate. CpG island methylation is a common aberration frequently associated with gene silencing in multiple tumor types, emerging as a highly promising biomarker. The transmembrane protein with a single EGF-like and two follistatin domains (TMEFF2) is epigenetically silenced in numerous tumor types, suggesting a potential role as a potential tumor suppressor. However, the role of TMEFF2 in lung cancer remains to be fully elucidated. We explored the methylation status of TMEFF2 gene in 139 patients with non-small cell lung cancer (NSCLC) and the feasibility of detecting circulating methylated DNA as a screening tool for NSCLC using methylation-specific PCR in 316 patients and 50 age-matched health controls. TMEFF2 methylation in tumor tissues was found in 73 of the 139 NSCLCs (52.5%) and was related to gene expression. The frequency of TMEFF2 methylation was higher in females and never-smokers than in males and smokers with borderline significance (65.8% vs 47.8%, p = 0.06; 65.7% vs 48.1%, p = 0.07). Notably, in adenocarcinomas, TMEFF2 methylation was significantly more frequent in tumors without EGFR mutation than those with EGFR mutation (adjusted odds ratio = 7.13, 95% confidence interval = 2.05-24.83, P = 0.002). Furthermore, TMEFF2 methylation was exclusively detected in the serum of NSCLC patients at a frequency of 9.2% (29/316). These findings suggest that methylation-associated down-regulation of TMEFF2 gene may be involved in lung tumorigenesis and TMEFF2 methylation can serve as a specific blood-based biomarker for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , DNA, Neoplasm/genetics , Lung Neoplasms/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Aged , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Culture Techniques , CpG Islands , DNA, Neoplasm/blood , DNA, Neoplasm/metabolism , Epigenomics , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Promoter Regions, Genetic
6.
PLoS One ; 7(6): e38974, 2012.
Article in English | MEDLINE | ID: mdl-22685614

ABSTRACT

Nuclear targeting of bacterial proteins has emerged as a pathogenic mechanism whereby bacterial proteins induce host cell pathology. In this study, we examined nuclear targeting of Acinetobacter baumannii transposase (Tnp) and subsequent epigenetic changes in host cells. Tnp of A. baumannii ATCC 17978 possesses nuclear localization signals (NLSs), (225)RKRKRK(230). Transient expression of A. baumannii Tnp fused with green fluorescent protein (GFP) resulted in the nuclear localization of these proteins in COS-7 cells, whereas the truncated Tnp without NLSs fused with GFP were exclusively localized in the cytoplasm. A. baumannii Tnp was found in outer membrane vesicles, which delivered this protein to the nucleus of host cells. Nuclear expression of A. baumannii Tnp fused with GFP in A549 cells induced DNA methylation of CpG regions in the promoters of E-cadherin (CDH1) gene, whereas the cytoplasmic localization of the truncated Tnp without NLSs fused with GFP did not induce DNA methylation. DNA methylation in the promoters of E-cadherin gene induced by nuclear targeting of A. baumannii Tnp resulted in down-regulation of gene expression. In conclusion, our data show that nuclear traffic of A. baumannii Tnp induces DNA methylation of CpG regions in the promoters of E-cadherin gene, which subsequently down-regulates gene expression. This study provides a new insight into the epigenetic control of host genes by bacterial proteins.


Subject(s)
Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Cadherins/genetics , DNA Methylation , Promoter Regions, Genetic/genetics , Transposases/genetics , Acinetobacter baumannii/enzymology , Active Transport, Cell Nucleus , Animals , Bacterial Proteins/metabolism , Blotting, Western , COS Cells , Cadherins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chlorocebus aethiops , CpG Islands/genetics , Cytoplasm/metabolism , Down-Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Confocal , Mutation , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transposases/metabolism
7.
Mol Med Rep ; 5(1): 239-44, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21971684

ABSTRACT

Lung cancer is the major health problem and leading cause of cancer-related deaths worldwide owing to late diagnosis and poor prognosis. Aberrant promoter methylation is an important mechanism for silencing tumor-suppressor genes in cancer and a promising tool for the development of molecular biomarkers. Ras association domain family 1A (RASSF1A), a pivotal gatekeeper of cell cycle progression, is highly methylated in a wide range of human sporadic tumors, including lung cancer. However, no significant prognostic implications have been observed in most studies qualitatively analyzed by methylation-specific PCR (MSP). We found that the RASSF1A promoter was aberrantly methylated in 44.7 and 37.4% of the tumors by pyrosequencing (PS) and MSP methods, respectively. RASSF1A methylation evaluated by the two methods was more frequent in ever-smokers and tumors with TP53 mutation than in never-smokers and tumors without TP53 mutation, respectively. Univariate and multivariate analyses revealed that strong methylation was an unfavorable prognostic factor with stage I (adjusted HR, 2.25; 95% CI 1.03-4.90; P=0.003) and squamous cell carcinoma patients (adjusted HR=2.25, 95% CI 1.03-4.90, P=0.042). Taken together, these results suggested that quantitative PS could gain wider applications in clinical samples as a promising method for early detection screening and prognosis compared with MSP.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , DNA Methylation , Lung Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Polymerase Chain Reaction , Promoter Regions, Genetic , Sequence Analysis, DNA , Smoking , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
8.
Cancer Biomark ; 12(2): 73-9, 2012.
Article in English | MEDLINE | ID: mdl-23396252

ABSTRACT

Dickkopf-1 (DKK1) is known as a negative regulator of the Wnt signaling pathway, which plays a crucial role in carcinogenesis. However, aberrant expression and the role of DKK1 in human cancers remain controversial. To estimate the role of DKK1 and its prognostic potential in lung cancer, promoter methylation of DKK1 was evaluated in 139 primary non-small cell lung cancers (NSCLCs) by methylation-specific PCR and its association with clinical and prognostic parameters. DKK1 hypermethylation was detected in 48.9% of neoplastic lung tissues and was significantly more frequent in stage I than the more advanced stages II-IIIA (P=0.04). Additionally, patients with DKK1 methylation had a better overall survival than those with no methylation under univariate analysis. When stratified by clinicopathologic features, DKK1 methylation was significantly associated with a favorable survival in a subset of patients. The current findings suggested that DKK1 promoter methylation may be a tumor-associated event in the early stage of NSCLC and could also be useful prognostic indicator for NSCLC. Further work may clarify the molecular basis of DKK1 action in progression of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , Intercellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Disease Progression , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Promoter Regions, Genetic , Republic of Korea , Wnt Signaling Pathway
9.
Mol Cells ; 32(4): 343-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22038593

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide and is usually associated with a late diagnosis and a poor prognosis. Thymosin ß(10) (TMSB10) is a monomeric actin sequestering protein that regulates actin cytoskeleton organization. The aberrant TMSB10 expression has been implicated in the pathogenesis of human cancers. However, its role in carcinogenesis is still controversial. To better understand the role of TMSB10 in lung tumorigenesis and its regulatory mechanism, we examined the methylation status and expression of the TMSB10 gene in non-small cell lung cancers (NSCLCs) using methylation-specific PCR (MSP) and immunohistochemistry (IHC), respectively. MSP analysis showed that the TMSB10 promoter was already unmethylated in most tumor tissues and became demethylated in 20 (14.4%) of the 139 NSCLCs. TMSB10 hypomethylation was not significantly correlated with the clinicopathological features. IHC showed that the TMSB10 protein was strongly expressed in the cytoplasm of malignant cells and its overexpression was detected in 50.0% of the tumor tissues compared to normal tissues. TMSB10 overexpression was frequently observed in sqaumous cell carcinomas compared to adenocarcinomas with border line significance (P = 0.072). However, TMSB10 methylation status was not linked to its overexpression. Collectively, these results suggest that TMSB10 hypomethylation may be a frequent event in NSCLCs, but it may not be a common mechanism underlying TMSB10 overexpression. However, further studies with large numbers of patients are needed to confirm our findings.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Thymosin/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Methylation , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/physiopathology , Male , Neoplasm Staging , Promoter Regions, Genetic/genetics , Survival Analysis , Up-Regulation/drug effects
10.
Respirology ; 16(8): 1203-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21585618

ABSTRACT

BACKGROUND AND OBJECTIVE: The exact role of the cystic fibrosis transmembrane conductance regulator (CFTR) in pathophysiology, and the mechanisms regulating its expression are poorly understood. The CFTR gene is known to be genetically or epigenetically associated with several cancers. In the present study, the methylation status of the promoter region of the CFTR gene and its expression in primary non-small cell lung cancer (NSCLC) were investigated. METHODS: The methylation status of the promoter region of the CFTR gene in NSCLC tissue was assessed by pyrosequencing and methylation-specific PCR. Expression of the CFTR gene was analysed by real-time PCR, and CFTR gene reactivation was investigated using 5-aza-2'-deoxycytidine. The correlation between methylation of the CFTR gene and the clinical features of the patients was assessed. RESULTS: Methylation of the CFTR gene in NSCLC was quantitatively high by pyrosequencing analysis and qualitatively frequent by methylation-specific PCR analysis. Expression of the CFTR gene was significantly lower in NSCLC compared with normal lung tissue. In addition, the demethylating agent 5-aza-2'-deoxycytidine increased CFTR gene expression. Methylation of the CFTR gene was significantly greater in squamous cell carcinomas than in adenocarcinomas. CFTR gene methylation was associated with significantly poorer survival in young patients, but not in elderly patients. CONCLUSIONS: These findings suggest that DNA methylation may be important for downregulation of CFTR gene expression in lung cancer. Promoter hypermethylation of the CFTR gene may be an important prognostic factor in younger patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Down-Regulation , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Republic of Korea/epidemiology
11.
Cancer ; 117(3): 590-6, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-20862745

ABSTRACT

BACKGROUND: Lung cancer is the leading cause of cancer-related deaths worldwide. Epigenetic inactivation of certain genes by aberrant promoter methylation is recognized as a crucial component in the initiation and progression of lung cancer. Response gene to complement 32 (RGC32) has been identified as a cell cycle regulator induced by activation of complements; however, its role in carcinogenesis is still controversial. METHODS: The authors examined the methylation status in the promoter region of RGC32 gene in nonsmall cell lung cancers (NSCLCs) using a methylation-specific PCR and correlated the results with clinicopathological features. RESULTS: RGC32 methylation was found in 45 of 173 NSCLCs (26.0%) and was related to the gene expression. RGC32 methylation was more frequent in females than in males (P<0.05). RGC32 methylation was not significantly associated with the prognosis of patients; however, when the patients were categorized by TP53 mutational status, the effect of RGC32 methylation on prognosis was significantly different between those with and without TP53 mutations (P = .005 [test for homogeneity]). Specifically, RGC32 methylation was associated with significantly worse survival in the cases with wild-type TP53, whereas it exhibited a better survival outcome in the cases with TP53 mutations. CONCLUSIONS: The current findings suggest that methylation-associated down-regulation of RGC32 plays an important role in the pathogenesis of NSCLC, particularly in females. However, further studies with a large number of cases are needed to confirm the authors' findings.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Proteins/genetics , DNA Methylation , Lung Neoplasms/genetics , Muscle Proteins/genetics , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Carcinoma, Non-Small-Cell Lung/mortality , Down-Regulation , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged
12.
Mol Cells ; 30(1): 89-92, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20652500

ABSTRACT

The growth arrest DNA-damage-inducible protein 45 (GADD45) can serve as a key coordinator of the stress response by regulating cell cycle progression, genomic stability, DNA repair, and other stress-related responses. Although deregulation of GADD45 expression has been reported in several types of human tumors, its role in lung cancer is still unknown. DNA hypermethylation of promoter CpG islands is known to be a major mechanism for epigenetic inactivation of tumor suppressor genes. We investigated the methylation status of GADD45 family genes (GADD45A, B, and G) in 139 patients with non-small cell lung cancer (NSCLC) using methylation-specific PCR (MSP) and correlated the results with clinicopathologic features of the patients. Methylation frequencies in tumors were 1.4% for GADD45A, 7.2% for GADD45B, and 31.6% for GADD45G. RT-PCR and MSP analysis showed that promoter methylation of the GADD45G gene resulted in downregulation of its mRNA expression. GADD45G methylation was significantly more frequent in female patients than male patients (P = 0.035). This finding suggests that methylation-associated down-regulation of the GADD45G gene may be involved in lung tumorigenesis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA Methylation , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Aged , CpG Islands , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
13.
Cancer Genet Cytogenet ; 197(1): 39-45, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20113835

ABSTRACT

Retinoid X receptors (RXRs) are nuclear receptors for retinoids that play a critical role in the regulation of growth and differentiation in normal and tumor cells. Deregulation of RXR expression has been reported in non-small cell lung cancer (NSCLC); however, the mechanism underlying the impaired expression of RXRs in lung cancer is not known. Aberrant methylation of promoter CpG islands is known to be a major mechanism for inactivation of tumor suppressor genes. We investigated the methylation status of the RXR genes in 139 surgically resected NSCLCs and correlated the results with the clinicopathologic characteristics of the patients. Methylation in the tumors was detected in all three genes: RXRA, 5.7%; RXRB, 4.3%; RXRG, 23.7%. Reverse transcriptase-polymerase chain reaction analysis showed that RXRG methylation correlates with mRNA expression. Methylation of the RXRG gene was not significantly associated with the prognosis of patients. When the patients were categorized by smoking status, however, the effect of RXRG methylation on prognosis was significantly different between never- and ever-smokers (P=0.003, test for homogeneity). Specifically, RXRG methylation was associated with a significantly worse survival in never-smokers; a trend to better survival outcome was observed for ever-smokers, although not statistically significant. This finding suggests that methylation-associated downregulation of the RXRG gene may play a differential role in the carcinogenesis of NSCLCs according to smoking status, but further studies are needed to confirm this.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epigenesis, Genetic , Lung Neoplasms/genetics , Retinoid X Receptors/genetics , Adult , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , DNA Methylation , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
14.
Cancer Sci ; 101(2): 568-72, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20175786

ABSTRACT

CpG islands (CGIs) hypermethylation is implicated in the pathogenesis of many cancers, including lung cancer. The phosphate and tension homolog (PTEN) is a tumor suppressor that controls a variety of biological processes including cell proliferation, growth, migration, and death. The defects in PTEN regulation have a profound impact on carcinogenesis. Herein, we have examined the methylation status of the human PTEN gene in 137 primary non-small-cell lung cancers (NSCLCs) by using a methylation-specific PCR and correlated the results with clinicopathological features. Promoter methylation of the PTEN gene was observed in 5.1%, 2.9%, and 0.0% of three different CpG regions, which were localized at -1460 to -1263, -984 to -848, and -300 to -128 nucleotides upstream of the translation start site, respectively. Reverse transcription-PCR and immunohistochemical analysis showed the methylation of the CGI region at -984 to -848 correlated more accurately with PTEN expression. In addition, no significant correlation was found between PTEN methylation and clinicopathological factors, including the survival rates. These findings suggest that promoter methylation is not an important mechanism for PTEN deregulation in NSCLCs from Koreans.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , Lung Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Aged , CpG Islands , Female , Humans , Immunohistochemistry , Korea , Male , Middle Aged , PTEN Phosphohydrolase/analysis , Promoter Regions, Genetic , RNA, Messenger/analysis
15.
FEBS Lett ; 584(3): 605-11, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20004198

ABSTRACT

Obox genes are preferentially expressed in the ovary, testis and oocyte, and play important roles in many developmental processes. In this study, we report that Obox4 and Obox6 are expressed in mouse embryonic stem cells (mESCs) and that Obox4 regulates histone family gene expression in mESCs. Obox4 protein expressing mESCs formed colonies with a flattened and irregular morphology, and exhibited decreased expression levels of self-renewal related proteins, such as Oct4 and Sox2, as well as reduced alkaline phosphatase activity. The results of microarray analysis and siRNA mediated knockdown experiments suggest that Obox4 is an upstream regulator of the histone gene family.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Histones/metabolism , Homeodomain Proteins/metabolism , Alkaline Phosphatase/metabolism , Animals , DNA Methylation/genetics , Histones/genetics , Homeodomain Proteins/genetics , Immunoblotting , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , SOXB1 Transcription Factors/metabolism
16.
J Microbiol ; 47(5): 641-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19851738

ABSTRACT

Nuclear targeting of bacterial proteins is an emerging pathogenic mechanism whereby bacterial proteins can interact with nuclear molecules and alter the physiology of host cells. The fully sequenced bacterial genome can predict proteins that target the nuclei of host cells based on the presence of nuclear localization signal (NLS). In the present study, we predicted bacterial proteins with the NLS sequences from Klebsiella pneumoniae by bioinformatic analysis, and 13 proteins were identified as carrying putative NLS sequences. Among them, HsdM, a subunit of KpnAl that is a type I restriction-modification system found in K. pneumoniae, was selected for the experimental proof of nuclear targeting in host cells. HsdM carried the NLS sequences, (7)KKAKAKK(13), in the N-terminus. A transient expression of HsdM-EGFP in COS-1 cells exhibited exclusively a nuclear localization of the fusion proteins, whereas the fusion proteins of HsdM with substitutions in residues lysine to alanine in the NLS sequences, (7)AAAKAAA(13), were localized in the cytoplasm. HsdM was co-localized with importin o in the nuclei of host cells. Recombinant HsdM alone methylated the eukaryotic DNA in vitro assay. Although HsdM tested in this study has not been considered to be a virulence factor, the prediction of NLS motifs from the full sequenced genome of bacteria extends our knowledge of functional genomics to understand subcellular targeting of bacterial proteins.


Subject(s)
Bacterial Proteins/genetics , Cell Nucleus/chemistry , DNA Restriction-Modification Enzymes/analysis , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/pathogenicity , Nuclear Localization Signals , Bacterial Proteins/analysis , Computational Biology/methods , DNA Restriction-Modification Enzymes/genetics , Genes, Bacterial , Genes, Reporter , Genome, Bacterial , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Humans , Karyopherins/analysis , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics
17.
Mol Carcinog ; 48(12): 1109-15, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19554572

ABSTRACT

Promoter methylation is an important mechanism in gene silencing and is a key epigenetic event in cancer development. Homeobox A5 (HOXA5) is a master regulator of the morphogenesis and cell differentiation to be implicated as a tumor suppressor gene in breast cancer, but its role in lung cancer is still unknown. In this study, we have investigated the methylation status of the promoter region of the HOXA5 gene in nonsmall cell lung cancers (NSCLCs) using nested and standard methylation-specific PCR (MSP) and correlated the methylation status with clinicopathological features. With standard MSP analysis, HOXA5 methylation were found in 113 (81.3%) of 139 NSCLCs and 72 (51.8%) in their corresponding nonmalignant lung tissues. RT-PCR and immunohistochemical analysis showed that HOXA5 methylation correlates with gene expression. Moreover, in the patients with stage I disease, HOXA5 methylation was more frequent in smokers than in never-smokes (P = 0.01). There was no influence of HOXA5 methylation on survival in all NSCLCs or at stages II-IV. However, in the patients with stage I disease, HOXA5 methylation was associated with a borderline significantly worse survival (P = 0.09). These findings suggest that downregulation of the HOXA5 gene by aberrant promoter methylation occurs in the vast majority of NSCLCs and that it may play a role in the pathogenesis of NSCLC. Additional studies with larger sample sizes are required to evaluate the prognostic value of HOXA5 methylation in patients with stage I NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Lung Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
18.
Lung Cancer ; 65(2): 247-50, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19362748

ABSTRACT

Lung cancer is the leading cause of cancer deaths worldwide and is usually associated with late diagnosis and poor prognosis. Tumor-acquired methylation of the promoter CpG islands (CGIs) is an important mechanism for silencing tumor suppressor genes. The checkpoint kinase 2 (CHK2) is a tumor suppressor that plays a crucial role in regulating cell-cycle checkpoints and apoptosis following DNA damage. The methylation statuses of two CGIs, distal and proximal, of human CHK2 gene were determined in non-small cell lung cancers (NSCLCs) using a nested methylation-specific PCR and bisulfite sequencing. The methylation of distal CHK2 CGI was found in 39 (28.1%) of the 139 NSCLCs. Its frequency was significantly more frequent in squamous cell carcinomas than in adenocarcinomas (40.0% vs 19.0%, p=0.006) and was also higher in ever-smokers than in never-smokers with a borderline significance (31.7% vs 17.1%, p=0.071). RT-PCR analysis showed that the distal CGI methylation correlated with CHK2 mRNA expression. However, the methylation of the proximal CHK2 CGI is not specific to tumors and not related to gene expression. These results suggest that the down-regulation of CHK2 gene via distal CGI methylation may play a role in the pathogenesis of NSCLC, particularly squamous cell carcinoma. However, further studies with large numbers of patients are needed to confirm our findings.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Checkpoint Kinase 2 , CpG Islands/genetics , Down-Regulation , Female , Gene Expression , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
19.
Dev Genes Evol ; 218(10): 561-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18810488

ABSTRACT

To examine whether the epigenetic status of the human Oct4 promoter is similarly regulated in mouse cells, we engineered a human bacterial artificial chromosome to express green fluorescent protein under the control of the hOct4 promoter and stably integrated it into mouse embryonic stem cells (mESCs), NIH3T3, and 293T cells. The hOct4 promoter is unmethylated in mESCs and it undergoes methylation during retinoic acid-induced differentiation. However, the hOct4 promoter is demethylated in NIH3T3 cells even though it is fully methylated in 293T cells. Methylation status of the hOct4 promoter is associated with green fluorescent protein expression at transcription level. Our findings indicate that the hOct4 promoter is differently regulated in mouse cells.


Subject(s)
Epigenesis, Genetic , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , Cell Line , Chromosomes, Artificial, Bacterial/genetics , DNA Methylation , Gene Expression Regulation, Developmental , Genetic Engineering , Green Fluorescent Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Organ Specificity , Plasmids/genetics
20.
Mol Cells ; 24(3): 364-71, 2007 Dec 31.
Article in English | MEDLINE | ID: mdl-18182852

ABSTRACT

The tumor suppressor gene Ras association domain family 1A (RASSF1A) is highly methylated in a wide range of human sporadic tumors. The current study investigated the hypermethylation of RASSF1A, the expression of RASSF1A protein, and the correlation between these and the clinicopathological features of gallbladder (GB) cancer in Korean patients. Formalin-fixed, paraffin-embedded tumors and non-neoplastic GB tissues (22 carcinomas, 8 adenomas, 26 normal epithelia) were collected from patients who had undergone surgical resection. The methylation status of two regions of the RASSF1A CpG island was determined by methylation-specific PCR (MSP), and the expression of RASSF1A protein was examined by immunohistochemistry using tissue microarrays. The K-RAS mutation was analyzed by direct sequencing. Methylation of the RASSF1A promoter (region 1) was detected in 22.7% (5/22) of carcinomas, 12.5% (1/8) of adenomas, and 0% (0/26) of normal gallbladder epithelia (P = 0.025). Methylation of the first exon (region 2) was found in 36.4% (8/22) of carcinomas, 25.0% (2/8) of adenomas, and 8.0% (2/26) of normal gallbladder epithelia (P = 0.038). K-RAS mutations were present in 4.5% (1/22) of carcinomas and 25% (2/8) of adenomas. RASSF1A methylaton was not associated with clinicopathological factors or K-ras mutation. Reduction or loss of RASSF1A expression was observed in most methylated adenocarcinomas. Three RASSF1A-expressing human biliary tract cancer cell lines examined contained unmethylated promoters and exons 1. These results suggest that downregulation of RASSF1A expression by DNA hypermethylation may be involved in GB carcinogenesis.


Subject(s)
Gallbladder Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , DNA Methylation , Gallbladder/metabolism , Gallbladder Neoplasms/genetics , Genes, ras/genetics , Humans , Immunohistochemistry , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...