Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Guang Pu Xue Yu Guang Pu Fen Xi ; 36(9): 2802-6, 2016 Sep.
Article in Zh | MEDLINE | ID: mdl-30084599

ABSTRACT

Rapid classification of leather variety means important to product process control, trading process and market surveillance. There is no official detection standard on classification of leather variety for the present. By now the testers use organoleptic method, burning method, chemical dissolution method, microscope method, or combination of them, to give a convincing result. The testers are required to highly sufficiently experienced, and not influenced by subjective factors. It also costs too much time. For the purpose of this research, spectra of five common varieties of leather samples (full-grain leather, split leather, sheep leather, reborn leather and manmade leather) were collected from market. Discriminant analysis combined with pre-processing method, including multiplicative signal correction (MSC), standard normal variate (SNV), first derivative and second derivative were used to classify the spectra above. It shows that the above five varieties of leather overlapped seriously in the same space. But manmade leather can be easily distinguished from the other four leather varieties using rear spectra, with the misclassified percent of 1.2%. The last four leather varieties covered each other partly in the same space, classify of any two of them can reach a lower misclassified percent, about 1.3%~17.9%. Different pre-processing method affected the discriminantion model positively or negatively with no regularity. None of these pre-processing methods was found to give a positive effect in a stable and persistent way. It can be concluded that it is feasible to discriminate the common leather varieties by near infrared Spectroscopy. All of the samples were taken from the finish products in the market (eg, handbag, belt, leather coat), which were processed in different ways (eg. tanning, knurling, dyeing). The different processes of the samples could bring an unforeseeable influence to the model which may be reduced by some method, for example, increasing the number and variety of samples.

2.
BMC Cancer ; 14: 418, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24917186

ABSTRACT

BACKGROUND: B-cell lymphoma 6 (BCL6) protein, an evolutionarily conserved zinc finger transcription factor, showed to be highly expressed in various human cancers in addition to malignancies in the lymphoid system. This study investigated the role of BCL6 expression in breast cancer and its clinical significance in breast cancer patients. METHODS: Expression of BCL6 protein was assessed using in situ hybridization and immunohistochemistry in 127 breast cancer patients and 50 patients with breast benign disease as well as in breast cell lines. Expression of BCL6 was restored or knocked down in two breast cancer cell lines (MCF-7 and T47D) using BCL6 cDNA and siRNA, respectively. The phenotypic change of these breast cancer cell lines was assessed using cell viability MTT, Transwell invasion, colony formation, and flow cytometry assays and in a xenograft mice model. Luciferase reporter gene, immunoblot, and qRT-PCR were used to investigate the molecular events after manipulated BCL6 expression in breast cancer cells. RESULTS: BCL6 protein was highly expressed in breast cancer cell lines and tissue specimens and expression of BCL6 protein was associated with disease progression and poor survival of breast cancer patients. In vitro, the forced expression of BCL6 results in increased proliferation, anchorage-independent growth, migration, invasion and survival of breast cancer cell lines, whereas knockdown of BCL6 expression reduced these oncogenic properties of breast cancer cells. Moreover, forced expression of BCL6 increased tumor growth and invasiveness in a nude mouse xenograft model. At the gene level, BCL6 was a target gene of miR-339-5p. Expression of BCL6 induced expression of CXCR4 and cyclinD1 proteins. CONCLUSIONS: The current study demonstrated the oncogenic property of BCL6 in breast cancer and further study could target BCL6 as a novel potential therapeutic strategy for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Animals , Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-bcl-6 , RNA, Small Interfering , Signal Transduction/genetics
3.
Mol Carcinog ; 52 Suppl 1: E80-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23390063

ABSTRACT

A single-nucleotide polymorphism (rs2274223: A5780G:His1927Arg) in the phospholipase C epsilon gene (PLCϵ) was recently identified as a susceptibility locus for esophageal cancer in Chinese subjects. To determine the underlying mechanisms of PLCϵ and this SNP in esophageal carcinogenesis, we analyzed PLCϵ genotypes, expression, and their correlation in esophageal cancer cell lines, non-transformed esophageal cells, 58 esophageal squamous cell carcinomas and 10,614 non-cancer subjects from China. We found that the G allele (AG or GG) was associated with increased PLCϵ mRNA and protein expression in esophageal cancer tissues and in esophageal cancer cell lines. G allele was also associated with higher enzyme activity, which might be associated with increased protein expression. Quantitative analysis of the C2 domain sequences revealed that A:G allelic imbalance was strongly linked to esophageal malignancy. Moreover, the analysis of 10,614 non-cancer subjects demonstrated that the G allele was strongly associated with moderate to severe esophagitis in the subjects from the high-incidence areas of China (OR 6.03, 95% CI 1.59-22.9 in high-incidence area vs. OR 0.74, 95% CI 0.33-1.64 in low-incidence area; P = 0.008). In conclusion, the PLCϵ gene, particularly the 5780G allele, might play a pivotal role in esophageal carcinogenesis via upregulating PLCϵ mRNA, protein, and enzyme activity, and augmenting inflammatory process in esophageal epithelium. Thus, 5780G allele may constitute a promising biomarker for esophageal squamous cell carcinoma risk stratification, early detection, and progression prediction.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Esophagitis/genetics , Phosphoinositide Phospholipase C/genetics , Polymorphism, Single Nucleotide/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Esophagitis/enzymology , Esophagitis/pathology , Genotype , Humans , Immunoblotting , Immunoenzyme Techniques , Neoplasm Staging , Phosphoinositide Phospholipase C/metabolism , Polymerase Chain Reaction , Prognosis , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured
4.
Environ Technol ; 34(17-20): 2853-8, 2013.
Article in English | MEDLINE | ID: mdl-24527650

ABSTRACT

Landfill leachate is posing an ever-greater environmental hazard. Recently, a process for purification combining activated carbon, microwave (MW) and Fenton oxidation has drawn much attention. In this study, the effectiveness of this process for the pretreatment of an old-age landfill leachate was tested. The effects of various parameters were investigated and the optimal condition included as follows: MW energy density, 6 W/mL; MW power, 300 W; radiation time, 8 min; H2O2 dosage, 0.1 mol/L; Fe(2+)-EDTA dosage, 0.02 mol/L; granular activated carbon (GAC) dosage, 6 g/L. Within the present experimental condition applied, the chemical oxygen demand (COD) removal reached 56.5%, and the ratio of 5-day biochemical oxygen demand to chemical oxygen demand (BOD5/COD) was enhanced from 0.122 to 0.462. Comparing with GAC, MW and Fenton alone or the combinations of any two of them, MW/Fenton/GAC displayed superior treatment efficiency. The MW/Fenton/GAC process is believed to be a promising pretreatment technology for biorefractory old-age landfill leachate.


Subject(s)
Charcoal/chemistry , Hydrogen Peroxide/chemistry , Iron/chemistry , Microwaves , Water Pollutants, Chemical/isolation & purification , Water Purification/methods , Oxidation-Reduction
5.
BMC Cancer ; 12: 51, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22292984

ABSTRACT

BACKGROUND: miRNAs, endogenous oligonucleotide RNAs, play an important role in mammary gland carcinogenesis and tumor progression. Detection of their expression and investigation of their functions could lead to discovery of novel biomarkers for breast cancer. METHODS: In situ hybridization was used to detect miR-133a expression in formalin-fixed paraffin-embedded breast surgical specimens from 26 benign, 34 pericancerously normal and 90 cancerous tissues. qRT-PCR was performed to assess miR-133a levels in 6 breast cell lines and 10 benign and 18 cancerous fresh breast tissue specimens. Cell viability, migration, and invasion assays were used to determine the role of miR-133a in regulation of breast cancer cell growth, migration, and invasion, respectively. Luciferase assay was performed to assess miR-133a binding to FSCN1 gene. RESULTS: Expression of miR-133a was reduced from normal through benign to cancerous breast tissues. Expression of miR-133a was also low in breast cancer cell lines. The reduced miR-133a expression was associated with lymph nodes metastasis, high clinical stages, and shorter relapse-free survivals of patients with breast cancer. Furthermore, transfection of miR-133a oligonucleotides slightly inhibited growth but significantly decreased migration and invasion capacity of breast cancer cells, compared with negative controls, whereas knockdown of miR-133a expression induced breast cancer cell migration and invasion. In addition, we identified a putative miR-133a binding site in the 3'-untranslated region (UTR) of Fascin1 (FSCN1) gene using an online bioinformatical tool. We found that miR-133a transfection significantly reduced expression of FSCN1 mRNA and protein. The luciferase reporter assay confirmed that FSCN1 was the direct target gene of miR-133a. CONCLUSIONS: miR-133a expression was lost in breast cancer tissues, loss of which was associated with lymph nodes metastasis, high clinical stages and shorter relapse-free survivals of patients with breast cancer. Functionally, miR-133a can suppress tumor cell invasion and migration and targeted the expression of FSCN1. Future study will verify whether detection of miR-133a expression can served as a novel biomarker for breast cancer progression and patient prognosis.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Proliferation , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/mortality , Carcinoma/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Survival , Cohort Studies , Female , Humans , Neoplasm Invasiveness/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis
6.
Int J Cancer ; 128(1): 132-43, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20309880

ABSTRACT

Altered microRNA (miRNA) expression has been found to promote carcinogenesis, but little is known about the role of miRNAs in esophageal cancer. In this study, we selected 10 miRNAs and analyzed their expression in 10 esophageal cancer cell lines and 158 tissue specimens using Northern blotting and in situ hybridization, respectively. We found that Let-7g, miR-21 and miR-195p were expressed in all 10 cell lines, miR-9 and miR-20a were not expressed in any of the cell lines, and miR-16-2, miR-30e, miR-34a, miR-126 and miR-200a were expressed in some of the cell lines but not others. In addition, transient transfection of miR-34a inhibited c-Met and cyclin D1 expression and esophageal cancer cell proliferation, whereas miR-16-2 suppressed RAR-ß(2) expression and increased tumor cell proliferation. Furthermore, we found that miR-126 expression was associated with tumor cell dedifferentiation and lymph node metastasis, miR-16-2 was associated with lymph node metastasis, and miR-195p was associated with higher pathologic disease stages in patients with esophageal adenocarcinoma. Kaplan-Meier analysis showed that miR-16-2 expression and miR-30e expression were associated with shorter overall and disease-free survival in all esophageal cancer patients. In addition, miR-16-2, miR-30e and miR-200a expression were associated with shorter overall and disease-free survival in patients with esophageal adenocarcinoma; however, miR-16-2, miR-30e and miR-200a expression were not associated with overall or disease-free survival in squamous cell carcinoma patients. Our data indicate that further evaluation of miR-30e and miR-16-2 as prognostic biomarkers is warranted in patients with esophageal adenocarcinoma. In addition, the role of miR-34a in esophageal cancer also warrants further study.


Subject(s)
Esophageal Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Blotting, Northern , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , In Situ Hybridization , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Male , Middle Aged , Multivariate Analysis , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
7.
Cancer ; 117(13): 2842-52, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21692045

ABSTRACT

BACKGROUND: Different microRNAs have been shown to have oncogenic and tumor-suppressive functions in human cancers. Detection of their expression may lead to identifying novel markers for breast cancer. METHODS: The authors detected miR-340 expression in 4 human breast cell lines and then focused on its role in regulation of tumor cell growth, migration, and invasion and target gene expression. They then analyzed miR-340 expression in benign and cancerous breast tissue specimens. RESULTS: Endogenous miR-340 expression was down-regulated in the more aggressive breast cancer cell lines, which was confirmed in breast cancer tissue specimens by using quantitative real-time polymerase chain reaction. Further studies showed that induction of miR-340 expression was able to suppress tumor cell migration and invasion, whereas knockdown of miR-340 expression induced breast cancer cell migration and invasion. At the gene level, the authors identified c-Met as a direct miR-340 target to mediate cell migration and invasion through regulation of MMP-2 and MMP-9 expression. Ex vivo, loss of miR-340 expression was associated with lymph node metastasis, high tumor histological grade, clinical stage, and shorter overall survival of breast cancer as well as increased c-Met expression in breast cancer tissue specimens. CONCLUSIONS: miR-340 may play an important role in breast cancer progression, suggesting that miR-340 should be further evaluated as a novel biomarker for breast cancer metastasis and prognosis, and potentially a therapeutic target.


Subject(s)
Breast Neoplasms/genetics , Cell Movement , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/metabolism , Receptors, Growth Factor/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Lymphatic Metastasis/genetics , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Middle Aged , Neoplasm Staging , Polymerase Chain Reaction , Proto-Oncogene Proteins c-met/genetics , RNA, Small Interfering , Receptors, Growth Factor/genetics , Survival Rate
8.
BMC Cancer ; 11: 32, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21266059

ABSTRACT

BACKGROUND: Retinoid receptor-induced gene-1 (RRIG1) is a novel gene that has been lost in several types of human cancers. The aim of this study was to determine whether RRIG1 plays a role in breast cancer, such as in the suppression of breast cancer cell growth and invasion. METHODS: Immunohistochemistry was used to detect RRIG1 expression in breast tissue specimens. Gene transfection was used to restore or knock down RRIG1 expression in breast cancer cell lines for analysis of cell viability, colony formation, and migration/invasion potential. Reverse-transcription polymerase chain reaction and western blot assays were used to detect the changes in gene expression. The RhoA activation assay was used to assess RRIG1-induced inhibition of RhoA activity. RESULTS: The immunohistochemical data showed that RRIG1 expression was reduced in breast cancer tissues compared with normal and atypical hyperplastic breast tissues. RRIG1 expression was inversely correlated with lymph node metastasis of breast cancer but was not associated with the status of hormone receptors, such as estrogen receptor, progesterone receptor, or HER2. Furthermore, restoration of RRIG1 expression inhibited proliferation, colony formation, migration, and invasion of breast cancer cells. Expression of RRIG1 also reduced phosphorylated Erk1/2 and Akt levels; c-Jun, MMP9, and Akt expressions; and RhoA activity. In contrast, knockdown of RRIG1 expression promoted breast cancer cell proliferation, colony formation, migration, and invasion potential. CONCLUSION: The data from the current study indicated that RRIG1 expression was reduced or lost in breast cancer and that restoration of RRIG1 expression suppressed breast cancer cell growth and invasion capacity. Future studies will determine the underlying molecular mechanisms and define RRIG1 as a tumor-suppressor gene in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA, Antisense/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Membrane Proteins/genetics , Middle Aged , Neoplasm Invasiveness , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
9.
Mol Cancer ; 9: 93, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20426865

ABSTRACT

Tobacco smoke is an important risk factor for various human cancers, including esophageal cancer. How benzo [a]pyrene diol epoxide (BPDE), a carcinogen present in tobacco smoke as well as in environmental pollution, induces esophageal carcinogenesis has yet to be defined. In this study, we investigated the molecular mechanism responsible for BPDE-suppressed expression of retinoic acid receptor-beta2 (RAR-beta2) in esophageal cancer cells. We treated esophageal cancer cells with BPDE before performing methylation-specific polymerase chain reaction (MSP) to find that BPDE induced methylation of the RAR-beta2 gene promoter. We then performed chromatin immunoprecipitation (ChIP) assays to find that BPDE recruited genes of the methylation machinery into the RAR-beta2 gene promoter. We found that BPDE recruited DNA (cytosine-5-)-methyltransferase 3 alpha (DNMT3A), but not beta (DNMT3B), in a time-dependent manner to methylate the RAR-beta2 gene promoter, which we confirmed by reverse transcription-polymerase chain reaction (RT-PCR) analysis of the reduced RAR-beta2 expression in these BPDE-treated esophageal cancer cell lines. However, BPDE did not significantly change DNMT3A expression, but it slightly reduced DNMT3B expression. DNA methylase inhibitor 5-aza-2'-deoxycytidine (5-Aza) and DNMT3A small hairpin RNA (shRNA) vector antagonized the effects of BPDE on RAR-beta2 expressions. Transient transfection of the DNMT3A shRNA vector also antagonized BPDE's effects on expression of RAR-beta2, c-Jun, phosphorylated extracellular signal-regulated protein kinases 1/2 (ERK1/2), and cyclooxygenase-2 (COX-2), suggesting a possible therapeutic effect. The results of this study form the link between the esophageal cancer risk factor BPDE and the reduced RAR-beta2 expression.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/toxicity , Carcinogens/toxicity , DNA (Cytosine-5-)-Methyltransferases/metabolism , Esophageal Neoplasms/metabolism , Gene Expression/drug effects , Receptors, Retinoic Acid/biosynthesis , Blotting, Northern , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Esophageal Neoplasms/genetics , Humans , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , DNA Methyltransferase 3B
10.
BMC Cancer ; 10: 542, 2010 Oct 09.
Article in English | MEDLINE | ID: mdl-20932331

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play an important role in the regulation of cell growth, differentiation, apoptosis, and carcinogenesis. Detection of their expression may lead to identifying novel markers for breast cancer. METHODS: We profiled miRNA expression in three breast cancer cell lines (MCF-7, MDA-MB-231, and MDA-MB-468) and then focused on one miRNA, miR-339-5p, for its role in regulation of tumor cell growth, migration, and invasion and target gene expression. We then analyzed miR-339-5p expression in benign and cancerous breast tissue specimens. RESULTS: A number of miRNAs were differentially expressed in these cancer cell lines. Real-time PCR indicated that miR-339-5p expression was downregulated in the aggressive cell lines MDA-MB-468 and MDA-MB-231 and in breast cancer tissues compared with benign tissues. Transfection of miR-339-5p oligonucleotides reduced cancer cell growth only slightly but significantly decreased tumor cell migration and invasion capacity compared with controls. Real-time PCR analysis showed that BCL-6, a potential target gene of miR-339-5p, was downregulated in MDA-MB-231 cells by miR-339-5p transfection. Furthermore, the reduced miR-339-5p expression was associated with an increase in metastasis to lymph nodes and with high clinical stages. Kaplan-Meier analyses found that the patients with miR-339-5p expression had better overall and relapse-free survivals compared with those without miR-339-5p expression. Cox proportional hazards analyses showed that miR-339-5p expression was an independent prognostic factor for breast cancer patients. CONCLUSIONS: MiR-339-5p may play an important role in breast cancer progression, suggesting that miR-339-5p should be further evaluated as a biomarker for predicting the survival of breast cancer patients.


Subject(s)
Breast Neoplasms/pathology , MicroRNAs/physiology , Apoptosis , Biomarkers, Tumor , Breast/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , DNA-Binding Proteins/metabolism , Disease Progression , Female , Humans , In Situ Hybridization , In Vitro Techniques , MicroRNAs/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Prognosis , Proto-Oncogene Proteins c-bcl-6 , Wound Healing
11.
Zhonghua Nan Ke Xue ; 16(7): 599-605, 2010 Jul.
Article in Zh | MEDLINE | ID: mdl-20873592

ABSTRACT

OBJECTIVE: Experimental evidence shows that microRNAs play an important role in the initiation and progression of human malignancies. The present study aimed to investigate the expressions of 6 microRNAs in prostate cancer (PCa) and their clinical significance. METHODS: We investigated the expression profiles of 6 microRNAs (let-7g, let-7d, miR-98, miR-96, miR-182 and miR-183) using the method of locked nucleic acid (LNA)-modified oligonucleotide in situ hybridization (ISH) and the technology of tissue microarray (TMA) with the formalin-fixed paraffin-embedded (FFPE) specimens from 52 patients with PCa and 38 with benign prostatic hyperplasia (BPH). Then we analyzed the correlation among the expressions of the 6 microRNAs in PCa and their correlation with the Gleason score and clinical stages of PCa. RESULTS: Compared with BPH, the PCa patients showed decreased expressions of miR-98, let-7d and let-7g, and decreased expressions of miR-96, miR-182 and miR-183, with statistically significant differences between the two groups (P < 0.05). The positive rate of the 6 microRNAs was significantly correlated with the Gleason grades of PCa (P < 0.05), but not with the age and serum PSA concentration of the patients (P > 0.05). The expressions of miR-96 and miR-182 were correlated with the clinical stages of the tumor (P < 0.05). There was a positive correlation among the expressions of miR-96, miR-182 and miR-183 (P = 0.00, r = 0.41), as well as between the expressions of let-7d and let-7g (P = 0.00, r = 0.46) in the PCa tissues. And the expression of miR-98 was positively correlated with those of let-7d and let-7g (P = 0.00, r = 0.46). CONCLUSION: The expression profiles of the microRNAs let-7d, let-7g, miR-98, miR-96, miR-182 and miR-183 reflect the biological behavior of PCa to some extent, and might be important biomarkers for the early detection and prognostic assessment of prostate cancer.


Subject(s)
Adenocarcinoma/metabolism , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/diagnosis , Aged , Aged, 80 and over , Biomarkers, Tumor , Gene Expression Profiling , Humans , Male , MicroRNAs/classification , Middle Aged , Prognosis , Prostatic Neoplasms/diagnosis
12.
Methods Mol Biol ; 471: 335-60, 2009.
Article in English | MEDLINE | ID: mdl-19109788

ABSTRACT

Esophageal cancer is a significant worldwide health problem because of its poor prognosis and high incidence in certain parts of the world. Tobacco smoke and alcohol consumption are significant risk factors for esophageal squamous cell carcinoma, whereas frequent gastroesophageal reflux and subsequent inflammatory reactions play a role in causing the adenocarcinoma. Esophageal carcinogenesis involves multiple genetic alterations. A large body of knowledge has been generated regarding molecular alterations associated with esophageal carcinogenesis. These alterations include aberrant cell cycle control, DNA repair, cellular enzymes, growth factor receptors, and nuclear receptors. This chapter reviews the most frequent gene alterations and their correlation with risk factors as well as the prevention strategies in esophageal cancer.


Subject(s)
Esophageal Neoplasms/etiology , Esophageal Neoplasms/genetics , Alcohol Drinking/adverse effects , Esophageal Neoplasms/prevention & control , Gastroesophageal Reflux/complications , Gene Expression Regulation, Neoplastic , Humans , Mutation , Risk Factors , Smoking/adverse effects
13.
Cancer Res ; 67(4): 1589-93, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308098

ABSTRACT

We previously showed that induction of retinoid receptor-induced gene-1 (RRIG1) expression inhibited RhoA activation and tumor cell colony formation, invasion, and proliferation, and these effects are associated with the suppression of extracellular signal-regulated protein kinases 1 and 2 phosphorylation and cyclooxygenase-2 expression. To further elucidate its role in tumor cell growth, gene expression, and tumorigenesis, we determined RRIG1 expression in breast and esophageal tissue specimens and then stably transfected RRIG1 into a TE-8 esophageal squamous cell carcinoma (SCC) cell line. We found that RRIG1 was expressed in normal mammary glands (10 of 10) but not all ductal carcinoma in situ [11 of 19 (57.9%), P = 0.018] and invasive cancer [14 of 30 (46.7%), P = 0.0023] tissues. Similarly, RRIG1 was expressed in normal esophageal epithelium (22 of 22) but not all dysplastic [6 of 43 (14%), P = 0.0001] and SCC [50 of 122 (41%), P = 0.0001] tissues. Furthermore, RRIG1 expression correlated positively with tumor differentiation but inversely with lymph node metastasis of esophageal SCC. Finally, the stable transfection of RRIG1 inhibited esophageal SCC cell growth and the expression of extracellular signal-regulated protein kinases 1 and 2 and cell cycle-related genes (e.g., cyclin D1, phosphorylated Rb, and E2F). RRIG1-transfected sublines also inhibited tumor development in nude mice. The results of this study indicate that RRIG1 plays a role in suppressing tumorigenesis.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Membrane Proteins/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Adult , Aged , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Differentiation/physiology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Humans , Lymphatic Metastasis , Male , Membrane Proteins/genetics , Mice , Mice, Nude , Middle Aged , Transfection , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics
14.
Int J Cancer ; 122(9): 2050-6, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18172859

ABSTRACT

Tumor progression and metastasis contribute to the great majority of breast cancer deaths. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) are thought to be involved in tumor progression and metastasis. Thus, we determined whether the expression of MMP-9 and TIMP-1 is associated with prognosis in breast cancer patients. We measured serum MMP-9 and TIMP-1 by enzyme-linked immunosorbent assay in 60 breast cancer patients, 18 benign breast disease patients and 15 healthy controls. We also evaluated the expression of MMP-9 and TIMP-1 protein and mRNA in paraffin-embedded tumor tissues from the 60 breast cancer patients by immunohistochemistry and in situ hybridization. We then correlated serum and tissue levels of MMP-9 and TIMP-1 in breast cancer samples and their expression with patients' clinicopathologic characteristics. We found that serum levels of MMP-9 and TIMP-1 were significantly higher in breast cancer patients than in benign breast disease and in healthy controls. High serum levels of MMP-9 and TIMP-1 were associated with lymph node metastasis, higher tumor stage and lower relapse-free and overall survival (OS) rates. Compared to low expression, high tissue expression of MMP-9 protein was associated with lymph node metastasis and higher tumor stage; and high tissue expression of TIMP-1 was associated with a lower OS rate. Our findings suggest that MMP-9 and TIMP-1 may further be evaluated as biomarkers for predicting progression and prognosis of breast cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Matrix Metalloproteinase 9/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Adult , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Breast Diseases/enzymology , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , In Situ Hybridization , Lymphatic Metastasis , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/genetics , Middle Aged , Predictive Value of Tests , Prognosis , RNA, Messenger/metabolism , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Survival Analysis , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/genetics
15.
Cancer Epidemiol Biomarkers Prev ; 17(1): 88-94, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18199714

ABSTRACT

Statins are a class of low molecular weight drugs that inhibit the rate-limiting enzyme of the mevalonate pathway 3-hydroxy-3-methylglutaryl-CoA reductase. Statins have been approved and effectively used to control hypercholesterolemia in clinical setting. Recent study showed statin's antitumor activity and suggested a potential role for prevention of human cancers. In this study, we did cell viability, DNA fragmentation, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays to evaluate the action of statins on prostate cancer cells and used Western blotting and RhoA activation assay to investigate the underlying molecular mechanism of action. Our data showed that lovastatin and simvastatin effectively decreased cell viability in three prostate cancer cell lines (PC3, DU145, and LnCap) by inducing apoptosis and cell growth arrest at G(1) phase. Both lovastatin and simvastatin induced activation of caspase-8, caspase-3, and, to a lesser extent, caspase-9. Both statins suppressed expression of Rb, phosphorylated Rb, cyclin D1, cyclin D3, CDK4, and CDK6, but induced p21 and p27 expression in prostate cancer cells. Furthermore, lovastatin and simvastatin suppressed RhoA activation and c-JUN expression, but not cyclooxygenase-2 expression. Our data showed that the antitumor activity of statins is due to induction of apoptosis and cell growth arrest. The underlying molecular mechanism of statin's action is mediated through inactivation of RhoA, which in turn induces caspase enzymatic activity and/or G(1) cell cycle. Future studies should focus on examining statins and other apoptosis-inducing drugs (e.g., cyclooxygenase-2 inhibitors or curcumin) together to assess their efficacy in prevention of prostate cancer.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Prostatic Neoplasms/pathology , Simvastatin/pharmacology , Blotting, Western , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Tumor Cells, Cultured/drug effects
16.
Mol Cell Biol ; 25(3): 1054-69, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15657432

ABSTRACT

Retinoic acid (RA) is the ligand for nuclear RA receptors (RARs and RXRs) and is crucial for normal epithelial cell growth and differentiation. During malignant transformation, human bronchial epithelial cells acquire a block in retinoid signaling caused in part by a transcriptional defect in RARs. Here, we show that activation of c-Jun N-terminal kinase (JNK) contributes to RAR dysfunction by phosphorylating RARalpha and inducing degradation through the ubiquitin-proteasomal pathway. Analysis of RARalpha mutants and phosphopeptide mapping revealed that RARalpha residues Thr181, Ser445, and Ser461 are phosphorylated by JNK. Mutation of these residues to alanines prevented efficient ubiquitination of RARalpha and increased the stability of the protein. We investigated the importance of RARalpha phosphorylation by JNK as a mediator of retinoid resistance in lung cancer. Mice that develop lung cancer from activation of a latent K-ras oncogene had high intratumoral JNK activity and low RARalpha levels and were resistant to treatment with an RAR ligand. JNK inhibition in a human lung cancer cell line enhanced RARalpha levels, ligand-induced activity of RXR-RAR dimers, and growth inhibition by RA. These findings point to JNK as a key mediator of aberrant retinoid signaling in lung cancer cells.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/metabolism , 3T3 Cells , Animals , COS Cells , Chlorocebus aethiops , Genes, ras/genetics , HeLa Cells , Humans , Mice , Mice, Transgenic , Mutation/genetics , Phosphorylation , Retinoic Acid Receptor alpha , Signal Transduction/physiology , Signal Transduction/radiation effects , Tumor Cells, Cultured , Ubiquitin/metabolism , Ultraviolet Rays/adverse effects
17.
Cancer Res ; 66(14): 7111-8, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16849557

ABSTRACT

The expression of retinoic acid receptor beta2 (RAR-beta2) is frequently lost in various cancers and their premalignant lesions. However, the restoration of RAR-beta2 expression inhibits tumor cell growth and suppresses cancer development. To understand the molecular mechanisms responsible for this RAR-beta2-mediated antitumor activity, we did restriction fragment differential display-PCR and cloned a novel retinoid receptor-induced gene 1 (RRIG1), which is differentially expressed in RAR-beta2-positive and RAR-beta2-negative tumor cells. RRIG1 cDNA contains 2,851 bp and encodes a protein with 276 amino acids; the gene is localized at chromosome 9q34. Expressed in a broad range of normal tissues, RRIG1 is also lost in various cancer specimens. RRIG1 mediates the effect of RAR-beta2 on cell growth and gene expression (e.g., extracellular signal-regulated kinase 1/2 and cyclooxygenase-2). The RRIG1 protein is expressed in the cell membrane and binds to and inhibits the activity of a small GTPase RhoA. Whereas induction of RRIG1 expression inhibits RhoA activation and f-actin formation and consequently reduces colony formation, invasion, and proliferation of esophageal cancer cells, antisense RRIG1 increases RhoA activity and f-actin formation and thus induces the colony formation, invasion, and proliferation of these cells. Our findings therefore show a novel molecular pathway involving RAR-beta2 regulation of RRIG1 expression and RRIG1-RhoA interaction. An understanding of this pathway may translate into better control of human cancer.


Subject(s)
Esophageal Neoplasms/metabolism , Membrane Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Growth Processes , Cell Line, Tumor , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary/genetics , Esophageal Neoplasms/genetics , Humans , Membrane Proteins/genetics , Molecular Sequence Data , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Tumor Suppressor Proteins/genetics , rhoA GTP-Binding Protein/antagonists & inhibitors
18.
Cancer Lett ; 253(1): 14-24, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17188427

ABSTRACT

Retinoids, a group of structural and functional analogs of vitamin A, are known to regulate a large number of essential biological processes and to suppress carcinogenesis. The effects of retinoids are mainly mediated by nuclear retinoid receptors, which include retinoic acid receptors (RARs) and retinoid X receptors (RXRs). Each receptor has three subtypes (alpha, beta, and gamma) and each subtype has different isoforms. Retinoic acid receptor-beta (RAR-beta) has four isoforms that have different affinities to retinoids and different biological functions. Loss of expression of RAR-beta(2) during cancer development is associated with tumorigenesis and retinoid resistance; induction of its expression, on the other hand, can suppress carcinogenesis. Expression of another isoform, RAR-beta(4), is increased in various types of cancer. RAR-beta(4) transgenic mice develop hyperplasia and neoplasia in various tissues, and induction of RAR-beta(4) expression increases the growth of tumor cells that do not express RAR-beta(2). Future studies will focus on molecular pathways involving RAR-beta(2) and the role of RAR-beta(4) in cancer development.


Subject(s)
Neoplasms/metabolism , Receptors, Retinoic Acid/physiology , Base Sequence , Biomarkers/metabolism , Forecasting , Humans , Molecular Sequence Data , Neoplasms/prevention & control , Protein Isoforms/metabolism , Receptors, Retinoic Acid/genetics
19.
Cancer Res ; 65(8): 3462-9, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15833882

ABSTRACT

Retinoids and their derivatives can suppress the development of cancer in animals and in humans. We and others have shown that retinoid X receptor (RXR)-selective retinoids or "rexinoids" suppress the development of breast cancer in several animal models with minimal toxicity. LGD1069 (Bexarotene) is a potent RXR-selective retinoid with reduced toxicity compared with naturally occurring retinoids. In this study, we investigated the expression of LGD1069-modulated biomarkers. We previously did cDNA array analysis of LGD1069-treated breast cells using Affymetrix microarrays. These studies identified many LGD1069-regulated genes, one of which was cyclooxygenase-2 (COX-2). Because COX-2 inhibitors have been shown to prevent cancer in other model systems, we investigated whether LGD1069 inhibits the expression of COX-2 in mammary tissue and in normal human mammary epithelial cells (HMEC). In mouse mammary tumor virus-erbB2 mice treated with LGD1069, there was a marked decrease of COX-2 expression in both normal and malignant mammary tissues. The effect of LGD1069 on COX-2 expression was also investigated in normal human breast cells. COX-2 expression was markedly reduced by treatment with LGD1069 at the RNA and protein level in normal HMECs; LGD1069 suppressed COX-2 promoter activity. We also showed that LGD1069 inhibited activator protein (AP-1)-dependent transcription in these breast cells, and that suppression of COX-2 expression was due to sequestration of CBP/p300. These results from in vivo and in vitro studies suggest that LGD1069, an RXR-selective retinoid, inhibits COX-2 expression by suppression of COX-2 transcription in part through transrepression of the AP-1 transcription factor. Thus, RXR-selective retinoids that inhibit AP-1 activity and suppress COX-2 expression may be particularly promising drugs for breast cancer prevention. Furthermore, such RXR-selective retinoids may be most useful in combination with antiestrogens for more effective prevention of breast cancer in women at high risk of this disease.


Subject(s)
Anticarcinogenic Agents/pharmacology , Breast Neoplasms/prevention & control , Breast/drug effects , Retinoid X Receptors/metabolism , Tetrahydronaphthalenes/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Animals , Bexarotene , Breast/enzymology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Down-Regulation/drug effects , E1A-Associated p300 Protein , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Membrane Proteins , Mice , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Promoter Regions, Genetic/drug effects , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Substrate Specificity , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects , Transfection
20.
Oncogene ; 24(56): 8268-76, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16170369

ABSTRACT

Benzo[a]pyrene diol epoxide (BPDE, a carcinogen present in tobacco smoke and environmental pollution) has been shown to suppress retinoic acid receptor-beta2 (RAR-beta(2)) and induce cyclooxygenase-2 (COX-2) expression. Restoration of RAR-beta(2) inhibited growth and colony formation of esophageal cancer cells, which was correlated with COX-2 suppression. In this study, we investigated the molecular mechanisms for RAR-beta(2)-mediated suppression of COX-2 expression using BPDE as a tool. We found that BPDE-induced COX-2 expression was through inhibition of RAR-beta(2) and consequently, induction of epidermal growth factor receptor (EGFR), extracellular signal-regulated protein kinases 1/2 (Erk1/2) phosphorylation, and c-Jun expression. Esophageal cancer cells that do not express RAR-beta(2) did not respond to BPDE for induction of COX-2. BPDE was also unable to induce COX-2 expression after RAR-beta(2) expression was manipulated in these esophageal cancer cells. Furthermore, BPDE induced time-dependent methylation of RAR-beta(2) gene promoter in esophageal cancer cells. Transfection of RAR-beta(2) expression vector into esophageal cancer cells suppressed expression of EGFR, Erk1/2 phosphorylation, c-Jun, and COX-2. In addition, co-treatment of RAR-beta(2)-positive cells with BPDE and the MEK1/2 inhibitor U0126 caused little change in c-Jun and COX-2 expression. This study demonstrated that BPDE-suppressed expression of RAR-beta(2) results in COX-2 induction and restoration of RAR-beta(2) expression reduces COX-2 protein in esophageal cancer cells, thereby further supporting our previous finding that RAR-beta(2) plays an important role in suppressing esophageal carcinogenesis.


Subject(s)
7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide/pharmacology , Cyclooxygenase 2/biosynthesis , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/biosynthesis , Animals , Butadienes/pharmacology , Cell Line, Transformed , Cell Line, Tumor , Cyclooxygenase 2/genetics , DNA Methylation/drug effects , Enzyme Induction/drug effects , Enzyme Induction/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Extracellular Signal-Regulated MAP Kinases/genetics , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mice, Nude , Multigene Family/physiology , Nitriles/pharmacology , Phosphorylation , Promoter Regions, Genetic/drug effects , Receptors, Retinoic Acid/physiology , Transcription Factor AP-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL