Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Ginseng Res ; 43(4): 527-538, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31695561

ABSTRACT

BACKGROUND: Ginsenoside Rg1 was shown to exert ligand-independent activation of estrogen receptor (ER) via mitogen-activated protein kinase-mediated pathway. Our study aimed to delineate the mechanisms by which Rg1 activates the rapid ER signaling pathways. METHODS: ER-positive human breast cancer MCF-7 cells and ER-negative human embryonic kidney HEK293 cells were treated with Rg1 (10-12M, 10-8M), 17ß-estradiol (10-8M), or vehicle. Immunoprecipitation was conducted to investigate the interactions between signaling protein and ER in MCF-7 cells. To determine the roles of these signaling proteins in the actions of Rg1, small interfering RNA or their inhibitors were applied. RESULTS: Rg1 rapidly induced ERα translocation to plasma membrane via caveolin-1 and the formation of signaling complex involving linker protein (Shc), insulin-like growth factor-I receptor, modulator of nongenomic activity of ER (MNAR), ERα, and cellular nonreceptor tyrosine kinase (c-Src) in MCF-7 cells. The induction of extracellular signal-regulated protein kinase and mitogen-activated protein kinase kinase (MEK) phosphorylation in MCF-7 cells by Rg1 was suppressed by cotreatment with small interfering RNA against these signaling proteins. The stimulatory effects of Rg1 on MEK phosphorylation in these cells were suppressed by both PP2 (Src kinase inhibitor) and AG1478 [epidermal growth factor receptor (EGFR) inhibitor]. In addition, Rg1-induced estrogenic activities, EGFR and MEK phosphorylation in MCF-7 cells were abolished by cotreatment with G15 (G protein-coupled estrogen receptor-1 antagonist). The increase in intracellular cyclic AMP accumulation, but not Ca mobilization, in MCF-7 cells by Rg1 could be abolished by G15. CONCLUSION: Ginsenoside Rg1 exerted estrogenic actions by rapidly inducing the formation of ER containing signalosome in MCF-7 cells. Additionally, Rg1 could activate EGFR and c-Src ER-independently and exert estrogenic effects via rapid activation of membrane-associated ER and G protein-coupled estrogen receptor.

2.
Front Pharmacol ; 7: 128, 2016.
Article in English | MEDLINE | ID: mdl-27252650

ABSTRACT

Phytoestrogen has been proposed as an alternative to hormone replacement therapy, which has been demonstrated to promote a high risk of breast cancer. However, the effect of phytoestrogen on breast cancer development has not been fully understood. Bakuchiol is an active ingredient of a traditional Chinese herbal medicine Fructus Psoraleae, the dried ripe fruit of Psoralea corylifolia L. (Fabaceae). The in vitro and in vivo estrogenic activities and anti-breast cancer effects of bakuchiol have not been well-studied. We found that bakuchiol induced the GFP expression in transgenic medaka (Oryzias melastigma, Tg, Chg:GFP) dose-dependently (0-1 µg/ml), demonstrating its in vivo estrogenic activity. Low dose of bakuchiol (1 µg/ml) induced the cell proliferation and ERα expression in MCF-7 cells, which could be blocked by the anti-estrogen ICI 182780, suggesting the in vitro estrogenic activity of bakuchiol. Our data indicated that high doses of bakuchiol (>2 µg/ml) inhibited breast cancer cell growth, with a stronger anti-proliferative effect than resveratrol, a widely studied analog of bakuchiol. High doses of bakuchiol (4, 7, and 10 µg/ml) were used for the further in vitro anti-breast cancer studies. Bakuchiol induced ERß expression and suppressed ERα expression in MCF-7 cells. It also induced S phase arrest in both MCF-7 and MDA-MB-231 cells, which could be rescued by caffeine. Knock-down of p21 also marginally rescued S phase arrest in MCF-7 cells. The S phase arrest was accompanied by the upregulation of ATM, P-Cdc2 (Tyr15), Myt1, P-Wee1 (Ser642), p21 and Cyclin B1, suggesting that blocking of Cdc2 activation may play an important role in bakuchiol-induced S phase arrest. Furthermore, bakuchiol induced cell apoptosis and disturbed mitochondrial membrane potential in MCF-7 cells. The bakuchiol-induced apoptosis was associated with increased expression of Caspase family and Bcl-2 family proteins, suggesting that bakuchiol may induce apoptosis via intrinsic apoptotic pathway. The in vivo anti-breast cancer effect of bakuchiol was further proved in zebrafish (Danio rerio, wild-type AB) xenografts. 0.5 µg/ml of bakuchiol significantly reduced the MCF-7 cell mass in zebrafish xenografts. Overall, these results suggested the potential of using bakuchiol in HRT and breast cancer treatment.

3.
Cancer Chemother Pharmacol ; 76(6): 1199-207, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26553104

ABSTRACT

PURPOSE: Adjunct chemoradiation is offered to unresectable esophageal squamous cell carcinoma (ESCC) patients, while its use is limited in tumors with strong resistance. Oxygen carriers or anti-hypoxic drugs belong to an emerging class of regulators that can alleviate tumor hypoxia. METHODS: We investigate the potential use of a novel oxygen carrier YQ23 in sensitizing chemoresistant ESCC in a series of subcutaneous tumor xenograft models developed using ESCC cell lines with different strengths of chemosensitivities. RESULTS: Tumor xenografts were developed using SLMT-1 and HKESC-2 ESCC cell lines with different strengths of resistance to two chemotherapeutic drugs, 5-fluorouracil and cisplatin. More resistant SLMT-1 xenografts responded better to YQ23 treatment than HKESC-2, as reflected by the induced tumor oxygen level. YQ23 sensitized SLMT-1 xenografts toward 5-fluorouracil via its effect on reducing the level of a hypoxic marker HIF-1α. Furthermore, a derangement of tumor microvessel density and integrity was demonstrated with a concurrent decrease in the level of a tumor mesenchymal marker vimentin. Similar to the 5-fluorouracil sensitizing effect, YQ23 also enhanced the response of SLMT-1 xenografts toward cisplatin by reducing the tumor size and the number of animals with invasive tumors. Chemosensitive HKESC-2 xenografts were irresponsive to combined YQ23 and cisplatin treatment. CONCLUSIONS: In all, YQ23 functions selectively on chemoresistant ESCC xenografts, which implicates its potential use as a chemosensitizing agent for ESCC patients.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Hemoglobins/pharmacology , Xenograft Model Antitumor Assays , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Synergism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Fluorouracil/pharmacology , Humans , Male , Mice, Nude , Oxygen/metabolism , Tumor Burden/drug effects
4.
J Steroid Biochem Mol Biol ; 141: 104-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24508513

ABSTRACT

Recent studies indicated that both estren and Rg1 appear to be able to activate mitogen-activated protein kinase (MAPK) pathway in estrogen responsive cells. Rg1 could lead to MAPK activation through ligand-independent activation of estrogen receptor (ER), while estren could activate the Src-MAPK pathway in an ERE-independent manner. Thus, it is important to understand the mechanistic insights on the difference in transcriptional activation between estren and Rg1. The present study also addressed the differential abilities of Rg1 and estren in terms of the ability to activate ER and the ability to induce ER translocation in MCF-7 cells. Our data indicated that Rg1 could increase pS2 gene expression, and could recruit the co-activator steroid receptor co-activator-1 (SRC-1) to the pS2 promoter. Rg1 could also induce ERα nuclear translocation as well as ERα phosphorylation at Ser118 principally in the cytoplasm in MCF-7 cells. We deduced that estren induced ERE-dependent transcriptional activity and activated ERα at Ser118 occurred in the nucleus of MCF-7 cells. However, it was found to decrease pS2 gene expression and failed to induce the recruitment of SRC-1 to the pS2 promoter in MCF-7 cells. Our results suggest that the abilities of Rg1 and estren to regulate pS2 gene expression, to recruit co-activators as well as to induce sub-cellular distribution of ERα are dramatically different.


Subject(s)
Estrenes/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/physiology , Ginsenosides/pharmacology , Breast Neoplasms , Cell Nucleus/metabolism , Cell Proliferation , Estradiol/pharmacology , Estradiol/physiology , Estrogens/pharmacology , Female , Gene Expression/drug effects , Humans , MCF-7 Cells , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Response Elements , Transcription, Genetic , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
PLoS One ; 8(10): e78395, 2013.
Article in English | MEDLINE | ID: mdl-24167620

ABSTRACT

Nasopharyngeal carcinoma (NPC) is common among southern Chinese including the ethnic Cantonese population living in Hong Kong. Epstein-Barr virus (EBV) infection is detected in all undifferentiated type of NPC in this endemic region. Establishment of stable and latent EBV infection in premalignant nasopharyngeal epithelial cells is an early event in NPC development and may contribute to its pathogenesis. Immortalized primary nasopharyngeal epithelial cells represent an important tool for investigation of EBV infection and its tumorigenic potential in this special type of epithelial cells. However, the limited availability and small sizes of nasopharyngeal biopsies have seriously restricted the establishment of primary nasopharyngeal epithelial cells for immortalization. A reliable and effective method to immortalize primary nasopharyngeal epithelial cells will provide unrestricted materials for EBV infection studies. An earlier study has reported that Bmi-1 expression could immortalize primary nasopharyngeal epithelial cells. However, its efficiency and actions in immortalization have not been fully characterized. Our studies showed that Bmi-1 expression alone has limited ability to immortalize primary nasopharyngeal epithelial cells and additional events are often required for its immortalization action. We have identified some of the key events associated with the immortalization of primary nasopharyngeal epithelial cells. Efficient immortalization of nasopharyngeal epithelial cells could be reproducibly and efficiently achieved by the combined actions of Bmi-1 expression, activation of telomerase and silencing of p16 gene. Activation of MAPK signaling and gene expression downstream of Bmi-1 were detected in the immortalized nasopharyngeal epithelial cells and may play a role in immortalization. Furthermore, these newly immortalized nasopharyngeal epithelial cells are susceptible to EBV infection and supported a type II latent EBV infection program characteristic of EBV-infected nasopharyngeal carcinoma. The establishment of an efficient method to immortalize primary nasopharyngeal epithelial cells will facilitate the investigation into the role of EBV infection in pathogenesis of nasopharyngeal carcinoma.


Subject(s)
Epithelial Cells/virology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Nasal Cavity/virology , Nasal Mucosa/virology , Pharynx/virology , Carcinoma , Cell Line, Transformed , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epstein-Barr Virus Infections/pathology , Female , Humans , MAP Kinase Signaling System , Male , Nasal Cavity/metabolism , Nasal Cavity/pathology , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Pharynx/metabolism , Pharynx/pathology , Polycomb Repressive Complex 1/metabolism , Virus Latency
6.
Carcinogenesis ; 34(4): 750-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23239745

ABSTRACT

Mitotic progression of mammalian cells is tightly regulated by the E3 ubiquitin ligase anaphase promoting complex (APC)/C. Deregulation of APC/C is frequently observed in cancer cells and is suggested to contribute to chromosome instability and cancer predisposition. In this study, we identified Daxx as a novel APC/C inhibitor frequently overexpressed in prostate cancer. Daxx interacts with the APC/C coactivators Cdc20 and Cdh1 in vivo, with the binding of Cdc20 dependent on the consensus destruction boxes near the N-terminal of the Daxx protein. Ectopic expression of Daxx, but not the D-box deleted mutant (DaxxΔD-box), inhibited the degradation of APC/Cdc20 and APC/Cdh1 substrates, leading to a transient delay in mitotic progression. Daxx is frequently upregulated in prostate cancer tissues; the expression level positively correlated with the Gleason score and disease metastasis (P = 0.027 and 0.032, respectively). Furthermore, ectopic expression of Daxx in a non-malignant prostate epithelial cell line induced polyploidy under mitotic stress. Our data suggest that Daxx may function as a novel APC/C inhibitor, which promotes chromosome instability during prostate cancer development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Cadherins/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Instability , Mitosis , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenomatous Polyposis Coli Protein/metabolism , Antigens, CD , Cdc20 Proteins , Cell Cycle , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Line, Tumor , Co-Repressor Proteins , HEK293 Cells , HeLa Cells , Humans , Male , Molecular Chaperones , Mutation , Neoplasm Grading , Neoplasm Metastasis , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering
7.
Nanomedicine ; 8(2): 204-11, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21704592

ABSTRACT

Multidrug resistance (MDR) is a major clinical obstacle to the success of cancer chemotherapy. Here we developed a gold-doxorubicin (DOX) nanoconjugates system to overcome MDR. Gold nanoparticles (AuNPs) were first PEGylated as Au-PEG-NH(2), and DOX was then grafted onto AuNPs via a cleavable disulfide linkage (Au-PEG-SS-DOX). Confocal images revealed that the extent of intracellular uptake of Au-PEG-SS-DOX was greater than that of free DOX in the MDR cells, and inductively coupled plasma mass spectroscopy analysis further confirmed that AuNPs significantly increased the level of drug accumulation in MDR cells at a nanoparticles dose greater than 15 µM. The cytotoxicity study demonstrated that the Au-PEG-SS-DOX nanoconjugates system efficiently released the anticancer drug DOX and enhanced its cytotoxicity against MDR cancer cells. This study highlights the potential of using AuNPs for overcoming of MDR in cancer chemotherapy. FROM THE CLINICAL EDITOR: This study demonstrates that gold nanoparticles can be successfully applied to overcome MDR in cancer chemotherapy.


Subject(s)
Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Resistance, Multiple/drug effects , Gold/chemistry , Metal Nanoparticles/chemistry , Doxorubicin/chemistry , Drug Carriers/adverse effects , Gold/adverse effects , Hep G2 Cells , Humans , Metal Nanoparticles/adverse effects , Microscopy, Confocal , Molecular Structure , Polyethylene Glycols/chemistry
8.
ACS Nano ; 5(10): 7838-47, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-21905691

ABSTRACT

Lanthanide-doped upconversion nanoparticles (UCNPs) are considered promising novel near-infrared (NIR) bioimaging agents with the characteristics of high contrast and high penetration depth. However, the interactions between charged UCNPs and mammalian cells have not been thoroughly studied, and the corresponding intracellular uptake pathways remain unclear. Herein, our research work involved the use of a hydrothermal method to synthesize polyvinylpyrrolidone-coated UCNPs (UCNP-PVP), and then a ligand exchange reaction was performed on UCNP-PVP, with the help of polyethylenimine (PEI) and poly(acrylic acid) (PAA), to generate UCNP-PEI and UCNP-PAA. These polymer-coated UCNPs demonstrated good dispersibility in aqueous medium, had the same elemental composition and crystal phase, shared similar TEM and dynamic light scattering (DLS) size distribution, and exhibited similar upconversion luminescence efficiency. However, the positively charged UCNP-PEI evinced greatly enhanced cellular uptake in comparison with its neutral or negative counterparts, as shown by multiphoton confocal microscopy and inductively coupled plasma mass spectrometry (ICP-MS) measurements. Meanwhile, we found that cationic UCNP-PEI can be effectively internalized mainly through the clathrin endocytic mechanism, as revealed by colocalization, chemical, and genetic inhibitor studies. This study elucidates the role of the surface polymer coatings in governing UCNP-cell interactions, and it is the first report on the endocytic mechanism of positively charged lanthanide-doped UCNPs. Furthermore, this study provides important guidance for the development of UCNPs as specific intracellular nanoprobes, allowing us to control the UCNP-cell interactions by tuning surface properties.


Subject(s)
Erbium/chemistry , Fluorides/chemistry , Molecular Imaging/methods , Nanoparticles/chemistry , Polymers/chemistry , Polymers/metabolism , Ytterbium/chemistry , Yttrium/chemistry , Chlorpromazine/pharmacology , Clathrin/antagonists & inhibitors , Clathrin/deficiency , Clathrin/genetics , Coated Vesicles/drug effects , Coated Vesicles/metabolism , Endocytosis/drug effects , Endocytosis/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Ligands , Luminescent Measurements , Nanoparticles/toxicity , Polymers/toxicity , Surface Properties
9.
Acta Pharmacol Sin ; 32(1): 52-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151160

ABSTRACT

AIM: To investigate the effects of the cardiotonic steroid, ouabain, on cardiac differentiation of murine embyronic stem cells (mESCs). METHODS: Cardiac differentiation of murine ESCs was enhanced by standard hanging drop method in the presence of ouabain (20 µmol/L) for 7 d. The dissociated ES derived cardiomyocytes were examined by flow cytometry, RT-PCR and confocal calcium imaging. RESULTS: Compared with control, mESCs treated with ouabain (20 µmol/L) yielded a significantly higher percentage of cardiomyocytes, and significantly increased expression of a panel of cardiac markers including Nkx 2.5, α-MHC, and ß-MHC. The α1 and 2- isoforms Na(+)/K(+)-ATPase, on which ouabain acted, were also increased in mESCs during differentiation. Among the three MAPKs involved in the cardiac hypertrophy pathway, ouabain enhanced ERK1/2 activation. Blockage of the Erk1/2 pathway by U0126 (10 µmol/L) inhibited cardiac differentiation while ouabain (20 µmol/L) rescued the effect. Interestingly, the expression of calcium handling proteins, including ryanodine receptor (RyR2) and sacroplasmic recticulum Ca(2+) ATPase (SERCA2a) was also upregulated in ouabain-treated mESCs. ESC-derived cardiomyocyes (CM) treated with ouabain appeared to have more mature calcium handling. As demonstrated by confocal Ca(2+) imaging, cardiomyocytes isolated from ouabain-treated mESCs exhibited higher maximum upstroke velocity (P<0.01) and maximum decay velocity (P<0.05), as well as a higher amplitude of caffeine induced Ca(2+) transient (P<0.05), suggesting more mature sarcoplasmic reticulum (SR). CONCLUSION: Ouabain induces cardiac differentiation and maturation of mESC-derived cardiomyocytes via activation of Erk1/2 and more mature SR for calcium handling.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/cytology , Ouabain/pharmacology , Animals , Calcium/metabolism , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression/drug effects , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Signal Transduction
10.
Cell Cycle ; 9(13): 2679-85, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20581448

ABSTRACT

The eukaryotic RNA splicing machinery is dedicated to the daunting task of excising intronic sequences on the many nascent RNA transcripts in a cell, and in doing so facilitates proper translation of its transcriptome. Notably, emerging evidence suggests that RNA splicing may also play direct roles in maintaining genome stability. Here we report the identification of the RNA/DNA-binding protein SON as a component of spliceosome that plays pleiotropic roles during mitotic progression. We found that SON is essential for cell proliferation, and that its inactivation triggers a MAD2-dependent mitotic delay. Moreover, SON deficiency is accompanied by defective chromosome congression, compromised chromosome segregation and cytokinesis, which in turn contributes to cellular aneuploidy and cell death. In summary, our study uncovers a specific link between SON and mitosis, and highlights the potential of RNA processing as additional regulatory mechanisms that govern cell proliferation and division.


Subject(s)
DNA-Binding Proteins/metabolism , Mitosis , Spliceosomes/metabolism , Cell Survival , Cytokinesis , Gene Silencing , HeLa Cells , Humans , Minor Histocompatibility Antigens , Protein Binding , Spindle Apparatus/metabolism
11.
Chem Commun (Camb) ; 46(14): 2391-3, 2010 Apr 14.
Article in English | MEDLINE | ID: mdl-20309461

ABSTRACT

The emission intensity of a monocationic Tb or Eu(III) complex of a bis(1-azaxanthone) ligand is enhanced in the presence of serum albumin; the lanthanide complex stains dividing cells, allowing visualisation of mitotic chromosomes.


Subject(s)
Coordination Complexes/chemistry , DNA/chemistry , Terbium/chemistry , Europium/chemistry , HeLa Cells , Humans , Ligands , Luminescence , Microscopy, Fluorescence
12.
Int J Cancer ; 127(7): 1570-83, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20091869

ABSTRACT

Epstein-Barr virus (EBV) infection has been postulated to be an early event involved in the pathogenesis of nasopharyngeal carcinomas (NPC). The lack of representative premalignant nasopharyngeal epithelial cell system for EBV infection has hampered research investigation into the regulation and involvement of EBV infection in NPC pathogenesis. We have compared the efficiency of EBV infection in nasopharyngeal epithelial cells with different biological properties including immortalized, primary and cancerous nasopharyngeal epithelial cells. EBV infection could be achieved in all the nasopharyngeal epithelial cells examined with variable infection rate. TGF-beta effectively enhanced EBV infection into nasopharyngeal epithelial cells both in the immortalized and primary nasopharyngeal epithelial cells. Stable infection of EBV was achieved in a telomerase-immortalized nasopharyngeal epithelial cell line, NP460hTert. The expression pattern of EBV-encoded genes and biological properties of this EBV infected cell line on long-term propagation were monitored. The EBV-infected nasopharyngeal epithelial cells acquired anchorage-independent growth and exhibited invasive growth properties on prolonged propagation. A distinguished feature of this EBV-infected nasopharyngeal epithelial cell model was its enhanced ability to survive under growth factor and nutrient starvation. This was evidenced by the suppressed activation of apoptotic markers and sustained activation of pAkt of EBV-infected cells compared to control cells under nutrient starvation. Examination of cytokine profiles of EBV-infected NP460hTert cells to nutrient and growth factor deprivation revealed upregulation of expression of MCP-1 and GRO-alpha. The establishment of a stable EBV infection model of premalignant nasopharyngeal epithelial cells will facilitate research investigation into the pathogenic role of EBV in NPC development.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharynx/pathology , Animals , Cell Line , Chromosome Mapping , DNA Primers , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/pathology , Gene Expression Profiling , Herpesvirus 4, Human/isolation & purification , Humans , Karyotyping , Mice , Mice, Nude , Nasopharyngeal Neoplasms/epidemiology , Nasopharyngeal Neoplasms/genetics , Nasopharynx/virology , Phenotype , Receptors, CCR2/genetics , Receptors, Complement 3d/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Cancer Lett ; 293(1): 41-51, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20092939

ABSTRACT

Ectopic expression of viral oncoproteins disrupts cellular functions and limits the value of many existing immortalization models as models for carcinogenesis, especially for cancers without definitive viral etiology. Our newly established telomerase-immortalized human esophageal epithelial cell line, NE2-hTERT, retained nearly-diploid and non-tumorigenic characteristics, but exhibited genetic and genomic alterations commonly found in esophageal cancer, including progressive loss of the p16(INK4a) alleles, upregulation of anti-apoptotic proteins, epithelial-mesenchymal transition, whole-chromosome 7 gain and duplicated 5q arm. Our data also revealed a novel positive regulation of p16(INK4a) on cyclin D1. These findings probably represent early crucial events and mechanisms in esophageal carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Esophageal Neoplasms/genetics , Esophagus/physiology , Neoplasms, Squamous Cell/genetics , Telomerase/genetics , Animals , Cell Differentiation/genetics , Cell Growth Processes/genetics , Cell Line, Transformed , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Epithelial Cells/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagus/metabolism , Esophagus/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , Retinoblastoma Protein/biosynthesis , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Spectral Karyotyping , Telomerase/biosynthesis , Transfection
14.
J Inorg Biochem ; 104(1): 62-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19880187

ABSTRACT

An amphiphilic porphyrin appended with a Ru(II)-polypyridyl complex (Ru-P) showing a moderate two-photon absorption cross-section (178.0+/-26.8GM), high singlet oxygen quantum yield and rapid cellular uptake was synthesized. In vitro study using human nasopharyngeal carcinoma cells showed that Ru-P exhibited a strong two-photon induced fluorescence upon uptake, lysosomal localization and potent two-photon induced cytotoxicity. These results show that Ru-P, which was designed to enhance its cellular uptake, can potentially be used as an efficacious bifunctional two-photon tumor-imaging and photodynamic therapeutic agent despite its moderate two-photon absorption cross-section.


Subject(s)
Antineoplastic Agents/metabolism , Imaging, Three-Dimensional , Neoplasms/metabolism , Organometallic Compounds/metabolism , Photochemotherapy , Photosensitizing Agents/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Neoplasms/drug therapy , Organometallic Compounds/chemistry , Organometallic Compounds/therapeutic use , Photons , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Spectrometry, Fluorescence
15.
16.
J Biophotonics ; 2(12): 718-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19685480

ABSTRACT

Highly emissive europium complexes with specific endoplasmic reticulum localization potential includes several advantages such as fast uptake, long resident lifetime, low dosage requirement, low cytotoxicity and highly emissive two-photon induced f-f emission imaging.


Subject(s)
Endoplasmic Reticulum/metabolism , Europium/analysis , Europium/chemistry , Luminescent Agents/metabolism , Organometallic Compounds/chemistry , Photons , Animals , Biological Transport , Cell Line , Endoplasmic Reticulum/drug effects , Europium/metabolism , Europium/toxicity , Humans , Luminescent Agents/analysis , Luminescent Agents/chemistry , Luminescent Agents/toxicity , Mice , Microscopy, Confocal , Organometallic Compounds/analysis , Organometallic Compounds/metabolism , Organometallic Compounds/toxicity , Staining and Labeling , Substrate Specificity
17.
Gene ; 443(1-2): 55-63, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19450668

ABSTRACT

RASSF1A, which is frequently found inactivated in human cancers, is revealed as a tumor suppressor gene in nasopharyngeal carcinoma (NPC). Using RASSF1A-expressing (NP69 and HK-1) and non-RASSF1A-expressing (C666-1) cell models, the transcriptional regulation of RASSF1A was studied. By deletion analysis of 3.1 kb of 5' flanking region, the core promoter of RASSF1A was identified in the region between -431 and -1 upstream of the translation start site. Sequence analysis of this core promoter revealed several putative transcription factor binding sties. Using NP69 cells and by block replacement mutagenesis, the presence of three functional GC-boxes were identified, to which by competitive and supershift electrophoretic mobility shift assays (EMSA), the in vitro bindings of Sp1 and Sp3 were suggested. The in vivo functions of Sp-proteins in regulating RASSF1A gene were then investigated by overexpression studies; among the tested Sp-proteins, Sp1 or Sp3, but not Sp4, was able to augment promoter activities. More interestingly, co-expression of Sp1 and Sp3 could synergistically enhance RASSF1A promoter function. UV irradiation induces oxidation stresses and hence is routinely used to investigate expressions of oncogenes and tumor suppressors. In this report, upon UV irradiation, the RASSF1A promoter activity and endogenous transcript levels were found to be reduced. By chromatin immunoprecipitation (ChIP) and EMSA, we demonstrated that the binding of Sp1 and Sp3 onto -431 to -202 were significantly reduced after UV irradiation. This UV-mediated effect on RASSF1A promoter, as shown by specific inhibitors that interrupt cellular pathways, is MEK1-, but not JNK-dependent. In summary, our data provided a simple model to explain the potential development of NPC, via silencing of the tumor suppressor RASSF1A by reduced bindings of activators Sp1 and Sp3 onto the GC-boxes in the core promoter of the gene.


Subject(s)
Down-Regulation , Nasopharyngeal Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , CpG Islands , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 4/metabolism , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Sp Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Ultraviolet Rays
18.
Inorg Chem ; 48(3): 872-8, 2009 Feb 02.
Article in English | MEDLINE | ID: mdl-19128152

ABSTRACT

The cyclometalated platinum(II) complex [Pt(L)Cl], where HL is a new cyclometalating ligand 2-phenyl-6-(1H-pyrazol-3-yl)pyridine containing C(phenyl), N(pyridyl), and N(pyrazolyl) donor moieties, was found to possess two-photon-induced luminescent properties. The two-photon-absorption cross section of the complex in N,N-dimethylformamide at room temperature was measured to be 20.8 GM. Upon two-photon excitation at 730 nm from a Ti:sapphire laser, bright-green emission was observed. Besides its two-photon-induced luminescent properties, [Pt(L)Cl] was able to be rapidly accumulated in live HeLa and NIH3T3 cells. The two-photon-induced luminescence of the complex was retained after live cell internalization and can be observed by two-photon confocal microscopy. Its bioaccumulation properties enabled time-lapse imaging of the internalization process of the dye into living cells. Cytotoxicity of [Pt(L)Cl] to both tested cell lines was low, according to MTT assays, even at loadings as high as 20 times the dose concentration for imaging for 6 h.


Subject(s)
Photons , Platinum Compounds/chemistry , Animals , Cell Line , Cell Survival/drug effects , Cyclization , Humans , Mice , Molecular Structure , Photochemistry , Platinum Compounds/toxicity , Spectrophotometry
19.
Mol Biol Cell ; 19(6): 2389-401, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18353975

ABSTRACT

The basic helix-loop-helix transcription factor, Id1, was shown to induce tetraploidy in telomerase-immortalized nasopharyngeal epithelial cells in this study. Using both transient and stable Id1-expressing cell models, multiple mitotic aberrations were detected, including centrosome amplification, binucleation, spindle defects, and microtubule perturbation. Many of these abnormal phenotypes have previously been reported in cells overexpressing Aurora A. Further experiments showed that Id1 could stabilize Aurora A, whereas knocking down Aurora A expression in Id1-expressing cells could rescue some of the mitotic defects. The mechanisms by which Aurora A could be modulated by Id1 were explored. DNA amplification of the Aurora A locus was not involved. Id1 could only weakly activate the transcriptional activity of the Aurora A promoter. We found that Id1 overexpression could affect Aurora A degradation, leading to its stabilization. Aurora A is normally degraded from mitosis exit by the APC/C(Cdh1)-mediated proteasomal proteolysis pathway. Our results revealed that Id1 and Cdh1 are binding partners. The association of Id1 and Cdh1 was found to be dependent on the canonical destruction box motif of Id1, the increased binding of which may compete with the interaction between Cdh1 and Aurora A, leading to stabilization of Aurora A in Id1-overexpressing cells.


Subject(s)
Inhibitor of Differentiation Protein 1/metabolism , Mitosis , Polyploidy , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Anaphase-Promoting Complex-Cyclosome , Aurora Kinases , Cell Line , Cell Polarity , Centrioles/enzymology , Cytokinesis , Down-Regulation , Enzyme Stability , Gene Amplification , Humans , Inhibitor of Differentiation Protein 1/chemistry , Inhibitor of Differentiation Protein 1/deficiency , Interphase , Microtubules/enzymology , Mutation/genetics , Neoplasms/enzymology , Neoplasms/pathology , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/enzymology , Substrate Specificity , Transcriptional Activation , Ubiquitin-Protein Ligase Complexes/metabolism , Up-Regulation
20.
J Am Chem Soc ; 130(12): 3714-5, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18321106

ABSTRACT

A polymeric terbium complex that can be excited by near-infrared excitation at 800 nm via multiphoton absorption processes has been synthesized. This complex has been demonstrated to show strong, observable, three-photon-induced f-f emission in cell imaging. In vitro studies carried out in three carcinoma cell lines (A549, HONE1, and HeLa) have been performed and shown to have low cytotoxicity. This complex is therefore a potential candidate for future infrared excitation imaging dyes.


Subject(s)
Carcinoma/metabolism , Fluorescent Dyes/chemistry , Image Enhancement/methods , Organometallic Compounds/chemistry , Polymers/chemistry , Terbium/chemistry , Cell Line, Tumor , Cell Membrane/chemistry , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Image Enhancement/instrumentation , Ligands , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Models, Molecular , Organometallic Compounds/chemical synthesis , Photons , Spectrometry, Fluorescence/instrumentation , Spectrometry, Fluorescence/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...