Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Immunol ; 133: 67-76, 2021 05.
Article in English | MEDLINE | ID: mdl-33636431

ABSTRACT

An imbalance in Th17 cells and Tregs may be an important cause of the pathogenesis of thymoma with myasthenia gravis (MG). In this study, 30 patients with simple thymoma and 30 patients with thymoma with MG were analyzed. Flow cytometry analysis of Th17 and Tregs in peripheral blood revealed that the percentages of Th17 in thymoma were lower than those in thymoma with MG, while the percentages of Tregs were higher than those in simple thymoma. Serum cytokine ELISA assays showed that IL-6 levels in simple thymoma were lower than those in MG patients. Further, Th17 and Tregs levels were detected by immunohistochemical double staining of thymoma tissue; the number of positive Th17 cells in thymoma with MG was higher than that in simple thymoma, while positive Tregs showed the opposite results. RORγt protein and mRNA expression in thymoma with MG were both higher than those in simple thymoma. FOXP3 protein and mRNA expression in the thymoma with MG group were lower than those in simple thymoma. The results of coculture of thymoma cells and CD4 + T cells showed that thymoma cells could promote the differentiation of Th17 cells and inhibit the Tregs. Overall, Th17 cells and related transcription factors and cytokines in thymoma with MG patients were higher than those in thymoma patients, whereas, Tregs showed the opposite results, the mechanism may be that thymoma can secrete IL6 and IL21. These findings indicated that imbalances in Th17/Tregs may account for the pathogeny of thymoma with MG.


Subject(s)
Myasthenia Gravis/immunology , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Thymoma/immunology , Adult , Aged , CD4 Lymphocyte Count , Cell Differentiation/immunology , Cell Line, Tumor , Coculture Techniques , Cytokines/blood , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Interleukin-6/metabolism , Interleukins/metabolism , Male , Middle Aged , Myasthenia Gravis/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA, Messenger/genetics , Thymoma/pathology , Transcription Factors/blood
2.
Oncol Lett ; 9(1): 341-346, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25435989

ABSTRACT

The present study examined the role of the PRDI-BF1-RIZ (PR) domain of tumor suppressor retinoblastoma protein-interacting zinc finger gene 1 (RIZ1) as an anticancer domain and its ability to induce apoptosis in esophageal squamous cell carcinoma (ESCC) cells. The TE13 ESCC cell line was transfected with pcDNA3.1(+) eukaryotic expression vectors bearing the open reading frames of either the human RIZ1 gene or the PR domain, and the mRNA and protein expression levels were then detected using quantitative reverse transcription polymerase chain reaction and western blotting, respectively. The rate of apoptosis was determined by flow cytometry and the cell invasion ability was determined by an invasion assay. RIZ1 and the PR domain induced apoptosis and reduced the cell invasion ability (P<0.01). These findings indicate that the RIZ1 gene possesses anticancer activity in the PR domain, which may be important in inhibiting the development of ESCC.

3.
Oncol Rep ; 32(3): 1064-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24993551

ABSTRACT

The present study aimed to investigate the expression and role of SET and MYND domain-containing protein 3 (SMYD3) in esophageal squamous cell carcinoma; to observe the proliferation of esophageal squamous cell carcinoma after suppression of SMYD3 expression; and to explore the effect of SMYD3 downregulation on the expression of retinoblastoma protein-interacting zinc finger gene 1 (RIZ1). Tissues from 11 patients, including cancer and normal esophageal tissues, were obtained by surgery to observe the SMYD3 protein expression immunohistochemistry. Esophageal squamous cell carcinoma TE13 cells were transfected with four different SMYD3-shRNA plasmids, and SMYD3 mRNA expression levels were assessed to select the most efficient interfering plasmid. After SMYD3 downregulation in TE13 cells, mRNA and protein expression levels of SMYD3 and RIZ1 were determined using RT-PCR and western blotting, and cell proliferation was evaluated by the MTT method. In all 11 tissue paired samples, SMYD3 protein expression was higher in the cancer tissues (72.7%; 8/11), than that in the normal tissues (18.2%; 2/11) (Fisher's exact test, P=0.03). The mRNA expression levels of SMYD3 were significantly decreased by RNA interference (P<0.05), and plasmid SMYD3-shRNA-1242 was determined to be the most effective. Compared with the controls, transfection with the SMYD3-shRNA interfering plasmid significantly reduced the SMYD3 mRNA and protein expression levels in TE13 cells (P<0.05), whereas the expression levels of the anti-oncogene RIZ1 were increased (P<0.05). The MTT assay showed that ablation of SMYD3 expression significantly inhibited proliferation of TE13 cells (P<0.05). SMYD3 may participate in the biological activity of esophageal squamous cell carcinoma, as overexpression of SMYD3 correlates with its occurrence and its downregulation inhibits cancer cell proliferation. The shRNA efficiently downregulated SMYD3 in TE13 cells, which represents an SMYD3-interfered cell-test-model for future experiments. RNAi suppression of SMYD3 promoted the expression of RIZ1 in TE13 cells, suggesting a signal transduction pathway between SMYD3 and RIZ1. The SMYD3-RIZ1 pathway may represent a therapeutic target for esophageal squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Esophageal Neoplasms/pathology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Humans , RNA Interference , Signal Transduction
4.
Oncotarget ; 5(10): 3386-98, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24930499

ABSTRACT

Although elevated expression of neogenin-1 has been detected in human gastric cancer tissue, its role in gastric tumorigenesis remains unclear due to the lack of neogenin-1 studies in cancer. Therefore, we demonstrated here the function and regulatory mechanism of neogenin-1 in gastric cancer. Neogenin-1 ablation decreased proliferation and migration of gastric cancer cells, whereas its over-expression reversed these effects. Xenografted analyses using gastric cancer cells displayed statistically significant inhibition of tumor growth by neogenin-1 depletion. Interestingly, galectin-3 interacted with HSF-1 directly, which facilitated nuclear-localization and binding on neogenin-1 promoter to drive its transcription and gastric cancer cell motility. The galectin-3-increased gastric cancer cell motility was down-regulated by HSF-1 depletion. Moreover, the parallel expression patterns of galectin-3 and neogenin-1, as well as those of HSF-1 and neogenin-1, were detected in the malignant tissues of gastric cancer patients. Taken together, high-expression of neogenin-1 promotes gastric cancer proliferation and motility and its expression is regulated by HSF-1 and galectin-3 interaction. In addition, we propose further studies for neogenin-1 and its associated pathways to provide them as a proper target for gastric cancer therapy.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Movement , Cell Proliferation , Membrane Proteins/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Galectin 3/metabolism , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors , Heterografts , Humans , Immunoprecipitation , Mice , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Transcription Factors/metabolism , Transfection , Up-Regulation
5.
World J Gastroenterol ; 19(37): 6170-7, 2013 Oct 07.
Article in English | MEDLINE | ID: mdl-24115813

ABSTRACT

AIM: To investigate the effect of retinoblastoma protein-interacting zinc finger gene 1 (RIZ1) upregulation in gene expression profile and oncogenicity of human esophageal squamous cell carcinoma (ESCC) cell line TE13. METHODS: TE13 cells were transfected with pcDNA3.1(+)/RIZ1 and pcDNA3.1(+). Changes in gene expression profile were screened and the microarray results were confirmed by reverse transcription-polymerase chain reaction (RT-PCR). Nude mice were inoculated with TE13 cells to establish ESCC xenografts. After two weeks, the inoculated mice were randomly divided into three groups. Tumors were injected with normal saline, transfection reagent pcDNA3.1(+) and transfection reagent pcDNA3.1(+)/RIZ1, respectively. Tumor development was quantified, and changes in gene expression of RIZ1 transfected tumors were detected by RT-PCR and Western blotting. RESULTS: DNA microarray data showed that RIZ1 transfection induced widespread changes in gene expression profile of cell line TE13, with 960 genes upregulated and 1163 downregulated. Treatment of tumor xenografts with RIZ1 recombinant plasmid significantly inhibited tumor growth, decreased tumor size, and increased expression of RIZ1 mRNA compared to control groups. The changes in gene expression profile were also observed in vivo after RIZ1 transfection. Most of the differentially expressed genes were associated with cell development, supervision of viral replication, lymphocyte costimulatory and immune system development in esophageal cells. RIZ1 gene may be involved in multiple cancer pathways, such as cytokine receptor interaction and transforming growth factor beta signaling. CONCLUSION: The development and progression of esophageal cancer are related to the inactivation of RIZ1. Virus infection may also be an important factor.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Gene Expression Profiling , Histone-Lysine N-Methyltransferase/genetics , Nuclear Proteins/genetics , Oncogenes , Transcription Factors/genetics , Animals , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factors/metabolism , Transfection , Tumor Burden , Up-Regulation
6.
Quant Imaging Med Surg ; 3(3): 132-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23833726

ABSTRACT

We have developed an indocyanine green-loaded perfluorocarbon (ICG/PFCE) nanoemulsion as a multifunctional theranostic nanomedicine which enables not only (19)F magnetic resonance (MR)/near-infrared fluorescence (NIRF) bimodal imaging but also subsequent photodynamic/photothermal dual therapy of cancer. The hydrodynamic size of ICG/PFCE nanoemulsions was 164.2 nm. The stability of indocyanine green (ICG) in aqueous solution was significantly improved when loaded on perfluorocarbon nanoemulsions. In addition, ICG/PFCE nanoemulsions showed good dispersion stability in aqueous media containing 10% fetal bovine serum, for at least 14 days. (19)F-MRI of ICG/PFCE nanoemulsions showed that the signal intensity increased with increasing nanoemulsion concentration with no signal observed from the surrounding background. Using NIRF imaging with perfluorocarbon nanoemulsion alone, without ICG, did not produce NIRF, while clear and bright fluorescent images were obtained with ICG/PFCE nanoemulsions at 10-µM ICG equivalent. The capacity of ICG-loaded nanoemulsions to generate heat following light irradiation by using an 810-nm laser was comparable to that of free ICG, while singlet oxygen generation of ICG-loaded nanoemulsions was significantly better than that of free ICG. In vitro cytotoxicity tests and fluorescence microscopy confirmed biocompatibility of the nanoemulsion. Upon light irradiation, U87MG glioblastoma cells incubated with ICG/PFCE nanoemulsions underwent necrotic cell death. The therapeutic mechanism during light illumination appears to be mainly due to the photodynamic effect at lower ICG concentrations, whilst the photothermal effect became more obvious at increased ICG concentrations, enabling combined photodynamic/photothermal therapy of cancer cells.

7.
Exp Mol Med ; 44(6): 387-93, 2012 Jun 30.
Article in English | MEDLINE | ID: mdl-22437631

ABSTRACT

Although mounting evidence indicates the involvement of galectin-3 in cancer progression and metastasis, the underlying molecular mechanisms remain largely unknown. In this study, we investigated the effect and possible mechanism of galectin-3 on the migration and invasion of B16F10, a metastatic melanoma cell line, in which galectin-3 and matrix metalloproteinase- 1 (MMP-1) were both found to be highly expressed. Knockdown of galectin-3 with specific siRNA reduced migration and invasion, which was associated with reduced expression of MMP-1. To further investigate the underlying mechanism, we examined the effect of galectin-3 knockdown on the activity of AP-1, a transcriptional factor regulating MMP-1 expression. We found that galectin-3 directly interacted with AP-1 and facilitated the binding of this complex to the MMP-1 promoter that drives MMP-1 transcription. Moreover, silencing of galectin-3 inhibited binding of fra-1 and c-Jun to promoter sites of MMP-1 gene. Consistent with these in vitro findings, our in vivo study demonstrated that galectin-3 shRNA treatment significantly reduced the total number of mouse lung metastatic nodules. Taken together, galectin- 3 facilitates cell migration and invasion in melanoma in vitro and can induce metastasis in vivo, in part through, regulating the transcription activity of AP-1 and thereby up-regulating MMP-1 expression.


Subject(s)
Galectin 3/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Transcription Factor AP-1/genetics , Animals , Binding Sites/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Galectin 3/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Metastasis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , RNA Interference , RNA, Small Interfering , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transcriptional Activation
8.
Clin Lab ; 58(1-2): 41-51, 2012.
Article in English | MEDLINE | ID: mdl-22372344

ABSTRACT

BACKGROUND: To study the expression of the RIZ1 (Retinoblastoma protein-interacting zinc-finger gene 1) gene and investigate the promoter region methylation status of RIZ1 gene in the human esophageal squamous cell carcinoma (ESCC) cell lines of KYSE150, KYSE510, TE13, EC9706, CaEsl7, and EC109. To investigate the influence of DNMT (DNA methyltransferase) 5-aza-CdR(5-aza-2'-deoxycytidine) on the transcription of the RIZ1 gene in one cell line whose RIZ1 gene promoter region methylation was detected, and to investigate its influence on the cell proliferation. METHODS: Real-time PCR (Real-time quantitative PCR) and an immunohistochemistry technique was used to get the expression of RIZ1 in specimens from 6 human ESCC cell lines and 28 ESCC patients (tumor tissues and adjacent non-cancerous tissues). MSP (Methylation-specific PCR) was used to investigate the promoter region methylation status of the RIZ1 gene in the 6 ESCC cell lines. One cell line, whose RIZ1 gene promoter region methylation was detected, was chosen for the next studies in which it was treated it by with 5-aza-CdR. Real-time PCR was used to investigate its influence on the transcription of RIZ1 gene and MTT (methyl thiazolyl tetrazolium) was used to detect if 5-aza-CdR inhibits the proliferation of the cell line. RESULTS: In the 28 ESCC patient samples, RIZ1 expression was significantly lower in the tumor tissues than that in their adjacent non-cancerous tissues (p < 0.05). Consistently, immunohistochemistry analyses of RIZ1 protein expression showed that in the ESCC tissues RIZ1 protein expression was also significantly lower than in the adjacent tissues. In the human ESCC tissues the rate of expression accounts for 0% (0/12), and in the adjacent noncancerous tissues the rate of expression was 66.7% (8/12), the correlation was highly significant (chi2 = 12.000, p < 0.05). Promoter methylation of the RIZ1 gene was detected in TE13, CaEsl7, EC109. The cell line TE13 was chosen for the next studies. The expression of RIZ1 mRNA in TE-13 was up-regulated after having been treated with 5-aza-CdR. 5-aza-CdR inhibited cell proliferation of TE-13 in a time and concentration-dependent manner. CONCLUSIONS: Promoter methylation may play an important role in the epigenetic silencing of RIZ1 gene expression. Methylation of the RIZ1 promoter and loss of RIZ1 expression in human ESCC are independent biomarkers. Their determination may offer guidance for selecting appropriate diagnoses and treatments. RIZ1 may be a potential tumor suppressor in human ESCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA Methylation/drug effects , DNA-Binding Proteins/metabolism , Decitabine , Drug Screening Assays, Antitumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , RNA, Messenger/metabolism , Transcription Factors/metabolism
9.
World J Gastroenterol ; 18(6): 576-82, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22363126

ABSTRACT

AIM: To investigate the promoter region methylation status of retinoblastoma protein-interacting zinc finger gene 1 (RIZ1) in the human esophageal squamous cell carcinoma (ESCC) cell lines and tissues and verify the relationship between methylation of RIZ1 and oncogenesis, tumor progression and metastasis etc of ESCC. METHODS: Methylation-specific polymerase chain reaction (MSP) was used to investigate the promoter region methylation status of RIZ1 in 6 ESCC cell lines. One cell line where RIZ1 promoter region methylation was detected was selected for the next study, where the cell line was treated with 5-aza-CdR. Real-time polymerase chain reaction was used to investigate its influence on the transcription of RIZ1. Experiments using frozen pathological specimens from 47 ESCC patients were performed using the same MSP methodology. RESULTS: Promoter methylation of RIZ1 gene was detected in TE13, CaEs17 and EC109 cell lines and the cell line TE13 was chosen for further study. The expression of RIZ1 mRNA in TE-13 was up-regulated after treatment with 5-aza-CdR. The rate of methylation in carcinomas tissues was significantly higher than those in matched neighboring normal and distal ending normal tissue, and the deviation of data was statistically significant (χ(2) = 24.136, P < 0.01). Analysis of the gender, age familial history, tumour deviation, tumour saturation, lymph gland displacement and clinical staging of 47 samples from ESCC patients showed that the fluctuation of data was not statistically significant. CONCLUSION: Promoter methylation may play an important role in the epigenetic silencing of RIZ1 gene expression in human ESCC. RIZ1 is considered to be a potential tumor suppressor gene and may be a biological parameter for testing early stage human ESCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Esophageal Neoplasms/genetics , Histone-Lysine N-Methyltransferase/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Base Sequence , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor/drug effects , Epigenesis, Genetic , Esophageal Neoplasms/pathology , Female , Gene Silencing , Humans , Male , Middle Aged , Molecular Sequence Data , Sequence Analysis, DNA
10.
Zhonghua Zhong Liu Za Zhi ; 27(8): 457-60, 2005 Aug.
Article in Chinese | MEDLINE | ID: mdl-16188139

ABSTRACT

OBJECTIVE: To investigate apoptosis-inducing effect and its mechanisms of HY-1, a carbazole alkaloid, on human erythroleukemia K562 cells. METHODS: Cell proliferation was detected by sulforhodamine B (SRB) assay after treated with HY-1 at indicated doses. Cell cycle analysis was performed by flow cytometry, mitochondria membrane voltage change was assessed by rhodamine 123 staining, annexin V-PI apoptosis detecting kit and DNA agarose gel electrophoresis were used to identify apoptosis-inducing effect of HY-1. The alterations of apoptosis-relating proteins were detected by Western blot. RESULTS: The IC(50) of HY-1 in K562 cells was (29.05 +/- 0.90) micromol/L by SRB assay. HY-1 had significant apoptotic inducing effect on K562 cells in a dose- and time-dependent manner as verified by appearance of Sub-G(1) peak on histogram of flow cytometry analysis, reduction of mitochondria membrane voltage, appearance of double positive cell group in Annexin V-PI apoptosis detecting test, and remarkable DNA ladder. The expression of cytosolic cytochrome c was apparently increased. Pro-caspase-9, pro-caspase-3 and PARP were all cleaved to active segments. There was no change in the expression of caspase-8. CONCLUSION: HY-1 exerts its anticancer activity through triggering apoptosis of K562 cells by mitochondria-activating pathways.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Rutaceae/chemistry , Alkaloids/isolation & purification , Alkaloids/pharmacology , Carbazoles/isolation & purification , Humans , K562 Cells , Mitochondria/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...