Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Int J Mol Sci ; 16(2): 2530-58, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25625511

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol, has been shown to inhibit the proliferation of a variety of tumor cells. Epidemiological studies have shown that drinking green tea can reduce the incidence of nasopharyngeal carcinoma (NPC), yet the underlying mechanism is not well understood. In this study, the inhibitory effect of EGCG was tested on a set of Epstein Barr virus-negative and -positive NPC cell lines. Treatment with EGCG inhibited the proliferation of NPC cells but did not affect the growth of a non-malignant nasopharyngeal cell line, NP460hTert. Moreover, EGCG treated cells had reduced migration and invasive properties. The expression of the cell adhesion molecules E-cadherin and ß-catenin was found to be up-regulated by EGCG treatment, while the down-regulation of matrix metalloproteinases (MMP)-2 and MMP-9 were found to be mediated by suppression of extracellular signal-regulated kinase (ERK) phosphorylation and AP-1 and Sp1 transactivation. Spheroid formation by NPC cells in suspension was significantly inhibited by EGCG. Oral administration of EGCG was capable of suppressing tumor growth in xenografted mice bearing NPC tumors. Treatment with EGCG was found to elevate the expression of p53 and p21, and eventually led to apoptosis of NPC cells via caspase 3 activation. The nuclear translocation of NF-κB and ß-catenin was also suppressed by EGCG treatment. These results indicate that EGCG can inhibit the proliferation and invasiveness, and induce apoptosis, of NPC cells, making it a promising agent for chemoprevention or adjuvant therapy of NPC.


Subject(s)
Apoptosis/drug effects , Catechin/analogs & derivatives , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Gelatinases/metabolism , Animals , Cadherins/metabolism , Carcinoma , Caspase 3/metabolism , Catechin/pharmacology , Catechin/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, SCID , NF-kappa B/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/enzymology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Transplantation, Heterologous , Up-Regulation/drug effects
2.
BMC Complement Altern Med ; 14: 300, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25127878

ABSTRACT

BACKGROUND: Abused women, who suffer from chronic psychological stress, have been shown to have shorter telomeres than never abused women. Telomere shortening is associated with increased risk of cell death, and it is believed that adopting health-promoting behaviors can help to increase the activity of telomerase, an enzyme that counters telomere shortening. Qigong is an ancient Chinese mind-body integration, health-oriented practice designed to enhance the function of qi, an energy that sustains well-being. Therefore, an assessor-blind, randomized, wait-list controlled trial was developed to evaluate the effect of a qigong intervention on telomerase activity (primary objective) and proinflammatory cytokines, perceived stress, perceived coping, and depressive symptoms (secondary objectives) in abused Chinese women. METHODS/DESIGN: A total of 240 Chinese women, aged ≥ 18 years, who have been abused by an intimate partner within the past three years will be recruited from a community setting in Hong Kong and randomized to receive either a qigong intervention or wait-list control condition as follows: the qigong intervention will comprise (i) a 2-hour group qigong training session twice a week for 6 weeks, (ii) a 1-hour follow-up group qigong exercise session once a week for 4 months, and (iii) a 30-minute self-practice qigong exercise session once a day for 5.5 months. The wait-list control group will receive qigong training after the intervention group completes the program. Upon completion of the qigong intervention program, it is expected that abused Chinese women in the intervention group will have higher levels of telomerase activity and perceived coping and lower levels of proinflammatory cytokines, perceived stress, and depressive symptoms than will abused Chinese women in the wait-list control group. DISCUSSION: This study will provide information about the effect of qigong exercise on telomerase activity and chronic psychological stress in abused Chinese women. The findings will inform the design of interventions to relieve the effects of IPV-related psychological stress on health. Also, the concept that health-promoting behaviors could slow down cellular aging might even motivate abused women to change their lifestyles. TRIAL REGISTRATION: Current Controlled Trials NCT02060123. Registered February 6, 2014.


Subject(s)
Battered Women , Qigong/methods , Stress, Psychological/therapy , Telomerase/blood , Waiting Lists , Adult , Female , Hong Kong , Humans , Randomized Controlled Trials as Topic/methods , Stress, Psychological/enzymology , Young Adult
3.
Mol Cancer ; 12(1): 128, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24156782

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is an epithelial malignancy strongly associated with Epstein-Barr virus (EBV). AT13387 is a novel heat shock protein 90 (Hsp90) inhibitor, which inhibits the chaperone function of Hsp90 and reduces expression of Hsp90-dependent client oncoproteins. This study aimed to evaluate both the in vitro and in vivo antitumor effects of AT13387 in the EBV-positive NPC cell line C666-1. RESULTS: Our results showed that AT13387 inhibited C666-1 cell growth and induced cellular senescence with the downregulation of multiple Hsp90 client oncoproteins EGFR, AKT, CDK4, and restored the protein expression of negative cell cycle regulator p27. We also studied the ability of AT13387 to restore p27 expression by downregulation of AKT and the p27 ubiquitin mediator, Skp2, using AKT inhibitor and Skp2 siRNA. In the functional study, AT13387 inhibited cell migration with downregulation of a cell migration regulator, HDAC6, and increased the acetylation and stabilization of α-tubulin. We also examined the effect of AT13387 on putative cancer stem cells (CSC) by 3-D tumor sphere formation assay. AT13387 effectively reduced both the number and size of C666-1 tumor spheres with decreased expression of NPC CSC-like markers CD44 and SOX2. In the in vivo study, AT13387 significantly suppressed tumor formation in C666-1 NPC xenografts. CONCLUSION: AT13387 suppressed cell growth, cell migration, tumor sphere formation and induced cellular senescence on EBV-positive NPC cell line C666-1. Also, the antitumor effect of AT13387 was demonstrated in an in vivo model. This study provided experimental evidence for the preclinical value of using AT13387 as an effective antitumor agent in treatment of NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Epstein-Barr Virus Infections/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoindoles/pharmacology , Nasopharyngeal Neoplasms/drug therapy , Acetylation , Animals , Carcinoma , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , Female , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Protein Processing, Post-Translational , Protein Stability , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tubulin/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Hepatology ; 55(2): 491-505, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953552

ABSTRACT

UNLABELLED: Emerging evidence implicates the chromodomain helicase/ATPase DNA binding protein 1-like gene (CHD1L) as a specific oncogene in human hepatocellular carcinoma (HCC). To better understand the molecular mechanisms underlying HCC cases carrying CHD1L amplification (>50% HCCs), we identified a CHD1L target, translationally controlled tumor protein (TCTP), and investigated its role in HCC progression. Here, we report that CHD1L protein directly binds to the promoter region (nt -733 to -1,027) of TCTP and activates TCTP transcription. Overexpression of TCTP was detected in 40.7% of human HCC samples analyzed and positively correlated with CHD1L overexpression. Clinically, overexpression of TCTP was significantly associated with the advanced tumor stage (P = 0.037) and overall survival time of HCC patients (P = 0.034). In multivariate analyses, TCTP was determined to be an independent marker associated with poor prognostic outcomes. In vitro and in vivo functional studies in mice showed that TCTP has tumorigenic abilities, and overexpression of TCTP induced by CHD1L contributed to the mitotic defects of tumor cells. Further mechanistic studies demonstrated that TCTP promoted the ubiquitin-proteasome degradation of Cdc25C during mitotic progression, which caused the failure in the dephosphorylation of Cdk1 on Tyr15 and decreased Cdk1 activity. As a consequence, the sudden drop of Cdk1 activity in mitosis induced a faster mitotic exit and chromosome missegregation, which led to chromosomal instability. The depletion experiment proved that the tumorigenicity of TCTP was linked to its role in mitotic defects. CONCLUSION: Collectively, we reveal a novel molecular pathway (CHD1L/TCTP/Cdc25C/Cdk1), which causes the malignant transformation of hepatocytes with the phenotypes of accelerated mitotic progression and the production of aneuploidy.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Chromosome Segregation , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , CDC2 Protein Kinase/metabolism , Cell Division , Cell Transformation, Neoplastic , Disease Progression , Electrophoresis, Gel, Two-Dimensional , G2 Phase , Hep G2 Cells , Humans , Tumor Protein, Translationally-Controlled 1 , Up-Regulation , cdc25 Phosphatases/metabolism
5.
Chin J Cancer ; 32(4): 205-12, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22704487

ABSTRACT

The discovery of induced pluripotent stem cells(iPSCs) is a promising advancement in the field of regenerative medicine. Previous studies have indicated that the teratoma-forming propensity of iPSCs is variable; however, the relationship between tumorigenic potential and genomic instability in human iPSCs (HiPSCs) remains to be fully elucidated. Here, we evaluated the malignant potential of HiPSCs by using both colony formation assays and tumorigenicity tests. We demonstrated that HiPSCs formed tumorigenic colonies when grown in cancer cell culture medium and produced malignancies in immunodeficient mice. Furthermore, we analyzed genomic instability in HiPSCs using whole-genome copy number variation analysis and determined that the extent of genomic instability was related with both the cells' propensity to form colonies and their potential for tumorigenesis. These findings indicate a risk for potential malignancy of HiPSCs derived from genomic instability and suggest that quality control tests, including comprehensive tumorigenicity assays and genomic integrity validation, should be rigorously executed before the clinical application of HiPSCs. In addition, HiPSCs should be generated through the use of combined factors or other approaches that decrease the likelihood of genomic instability.


Subject(s)
Carcinogenesis , DNA Copy Number Variations , Genomic Instability , Induced Pluripotent Stem Cells , Teratoma/etiology , Animals , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Mice , Mice, SCID , NIH 3T3 Cells , Octamer Transcription Factor-3/metabolism , Teratocarcinoma/etiology , Tumor Stem Cell Assay
6.
Nat Commun ; 14(1): 1912, 2023 04 06.
Article in English | MEDLINE | ID: mdl-37024479

ABSTRACT

Despite the intense CD8+ T-cell infiltration in the tumor microenvironment of nasopharyngeal carcinoma, anti-PD-1 immunotherapy shows an unsatisfactory response rate in clinical trials, hindered by immunosuppressive signals. To understand how microenvironmental characteristics alter immune homeostasis and limit immunotherapy efficacy in nasopharyngeal carcinoma, here we establish a multi-center single-cell cohort based on public data, containing 357,206 cells from 50 patient samples. We reveal that nasopharyngeal carcinoma cells enhance development and suppressive activity of regulatory T cells via CD70-CD27 interaction. CD70 blocking reverts Treg-mediated suppression and thus reinvigorate CD8+ T-cell immunity. Anti-CD70+ anti-PD-1 therapy is evaluated in xenograft-derived organoids and humanized mice, exhibiting an improved tumor-killing efficacy. Mechanistically, CD70 knockout inhibits a collective lipid signaling network in CD4+ naïve and regulatory T cells involving mitochondrial integrity, cholesterol homeostasis, and fatty acid metabolism. Furthermore, ATAC-Seq delineates that CD70 is transcriptionally upregulated by NFKB2 via an Epstein-Barr virus-dependent epigenetic modification. Our findings identify CD70+ nasopharyngeal carcinoma cells as a metabolic switch that enforces the lipid-driven development, functional specialization and homeostasis of Tregs, leading to immune evasion. This study also demonstrates that CD70 blockade can act synergistically with anti-PD-1 treatment to reinvigorate T-cell immunity against nasopharyngeal carcinoma.


Subject(s)
Epstein-Barr Virus Infections , Nasopharyngeal Neoplasms , Humans , Animals , Mice , T-Lymphocytes, Regulatory , Nasopharyngeal Carcinoma/genetics , CD27 Ligand/genetics , CD27 Ligand/metabolism , Herpesvirus 4, Human/metabolism , Nasopharyngeal Neoplasms/genetics , Lipids , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Tumor Microenvironment
7.
Hepatology ; 50(1): 122-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19441106

ABSTRACT

UNLABELLED: Amplification of 1q21 has been detected in 58% to 78% of primary hepatocellular carcinoma cases, suggesting that one or more oncogenes within the amplicon play a critical role in the development of this disease. The chromodomain helicase/adenosine triphosphatase DNA binding protein 1-like gene (CHD1L) is a recently identified oncogene localized at 1q21. Our previous studies have demonstrated that CHD1L has strong tumorigenic ability and confers high susceptibility to spontaneous tumors in a CHD1L-transgenic mouse model. In this study, we demonstrate that the antiapoptotic ability of CHD1L is associated with its interaction with Nur77, a critical member of a p53-independent apoptotic pathway. As the first cellular protein identified to bind Nur77, CHD1L is able to inhibit the nucleus-to-mitochondria translocation of Nur77, which is the key step of Nur77-mediated apoptosis, resulting in the hindrance of the release of cytochrome c and the initiation of apoptosis. Knock-down of CHD1L expression by RNA interference could rescue the mitochondrial targeting of Nur77 and the subsequent apoptosis. Further studies found that the C-terminal Macro domain of CHD1L is responsible for the interaction with Nur77, and a CHD1L mutant lacking residues 600-897 failed to interact with Nur77 and prevented Nur77-mediated apoptosis. More importantly, we found that the inhibition of Nur77-mediated apoptosis by endogenous CHD1L is a critical biological cellular process in hepatocarcinogenesis. CONCLUSION: We demonstrate in this study that overexpression of CHD1L could sustain tumor cell survival by preventing Nur77-mediated apoptosis.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Nucleus , DNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Mitochondria , Protein Transport , Receptors, Steroid/metabolism , Adenosine Triphosphatases/physiology , Cell Survival/genetics , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1
8.
Am J Cancer Res ; 10(6): 1710-1727, 2020.
Article in English | MEDLINE | ID: mdl-32642285

ABSTRACT

Oral tongue squamous cell carcinoma (OTSCC) has a distinctive cell sub-population known as tumor-initiating cells (TICs). CD271 is a functional TIC receptor in head and neck cancers. The molecular mechanisms governing CD271 up-regulation remains unclear. Oxidative stress is a contributing factor in TIC development. Here, we explored the potential role of NADPH oxidase 5 (NOX5) and its regulatory mechanism on the development of CD271-expressing OTSCC. Our results showed that the splice variant NOX5α is the most prevalent form expressed in head and neck cancers. NOX5α enhanced OTSCC proliferation, migration, and invasion. Overexpression of NOX5α increased the size of OTSCC xenograft significantly in vivo. The tumor-promoting functions of NOX5α were mediated through the reactive oxygen species (ROS)-generating property. NOX5α activated ERK singling and increased CD271 expression at the transcription level. Also, NOX5α reduces the sensitivity of OTSCC to cisplatin and natural killer cells. The findings indicate that NOX5α plays an important part in the development of TIC in OTSCC.

9.
Phytomedicine ; 63: 153058, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31394414

ABSTRACT

BACKGROUND: Sulforaphane (SFN), a natural compound present in cruciferous vegetable, has been shown to possess anti-cancer activities. Cancer stem cell (CSC) in bulk tumor is generally considered as treatment resistant cell and involved in cancer recurrence. The effects of SFN on nasopharyngeal carcinoma (NPC) CSCs have not yet been explored. PURPOSE: The present study aims to examine the anti-tumor activities of SFN on NPC cells with CSC-like properties and the underlying mechanisms. METHODS: NPC cells growing in monolayer culture, CSCs-enriched NPC tumor spheres, and also the NPC nude mice xenograft were used to study the anti-tumor activities of SFN on NPC. The population of cells expressing CSC-associated markers was evaluated using flow cytometry and aldehyde dehydrogenase (ALDH) activity assay. The effect of DNA methyltransferase 1 (DNMT1) on the growth of NPC cells was analyzed by using small interfering RNA (siRNA)-mediated silencing method. RESULTS: SFN was found to inhibit the formation of CSC-enriched NPC tumor spheres and reduce the population of cells with CSC-associated properties (SRY (Sex determining Region Y)-box 2 (SOX2) and ALDH). In the functional study, SFN was found to restore the expression of Wnt inhibitory factor 1 (WIF1) and the effect was accompanied with the downregulation of DNMT1. The functional activities of WIF1 and DNMT1 were confirmed using exogenously added recombinant WIF1 and siRNA knockdown of DNMT1. Moreover, SFN was found to inhibit the in vivo growth of C666-1 cells and enhance the anti-tumor effects of cisplatin. CONCLUSION: Taken together, we demonstrated that SFN could suppress the growth of NPC cells via the DNMT1/WIF1 axis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Isothiocyanates/pharmacology , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Neoplasms/drug therapy , Adaptor Proteins, Signal Transducing/genetics , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brassicaceae/chemistry , Cell Line, Tumor , Cisplatin/administration & dosage , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Humans , Isothiocyanates/administration & dosage , Male , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sulfoxides , Xenograft Model Antitumor Assays
10.
Cancer Lett ; 457: 98-109, 2019 08 10.
Article in English | MEDLINE | ID: mdl-31100412

ABSTRACT

ATP-binding cassette (ABC) transporters mediate multidrug resistance and cancer stem cell properties in various model systems. Yet, their biological significance in cancers, especially in hepatocellular carcinoma (HCC), remains unclear. In this study, we investigated the function of ABCF1 in HCC and explored its potential as a therapeutic target. ABCF1 was highly expressed in fetal mouse livers but not in normal adult livers. ABCF1 expression was upregulated in HCCs. These results demonstrate that ABCF1 functions as a hepatic oncofetal protein. We further demonstrated elevated ABCF1 expression in HCC cells upon acquiring chemoresistance. Suppression of ABCF1 by siRNA sensitized both parental cells and chemoresistant cells to chemotherapeutic agents. Reversely, ABCF1 overexpression promoted chemoresistance and drug efflux. In addition, overexpression of ABCF1 enhanced migration, spheroid and colony formation and epithelial-mesenchymal transition (EMT) induction. RNA sequencing analysis revealed EMT inducers HIF1α/IL8 and Sox4 as potential mediators for the oncogenic effect of ABCF1 in HCC progression. Together, this study illustrates that ABCF1 is a novel potential therapeutic target for HCC treatment.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Liver Neoplasms/metabolism , Neoplastic Stem Cells/drug effects , ATP-Binding Cassette Transporters/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Movement , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Inbred ICR , Mice, Nude , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
11.
Int J Oncol ; 54(3): 1010-1020, 2019 03.
Article in English | MEDLINE | ID: mdl-30569106

ABSTRACT

The Wnt signaling pathway is known to serve an important role in the control of cell migration. The present study analyzed the mechanisms underlying the in vitro modulation of the migration of nasopharyngeal carcinoma (NPC) cells by the CREB­binding protein/catenin antagonist and Wnt modulator ICG­001. The results revealed that ICG­001­mediated inhibition of tumor cell migration involved downregulated mRNA and protein expression of the Wnt target gene cluster of differentiation (CD)44. It was also demonstrated that ICG­001 downregulated the expression of CD44, and this effect was accompanied by restored expression of microRNA (miRNA)­150 in various NPC cell lines. Using a CD44 3'­untranslated region luciferase reporter assay, miR­150 was confirmed to be a novel CD44­targeting miRNA, which could directly target CD44 and subsequently regulate the migration of NPC cells. The present study provides further insight into the inhibition of tumor cell migration through the modulation of miRNA expression by the Wnt modulator ICG­001.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Movement/drug effects , Hyaluronan Receptors/genetics , MicroRNAs/genetics , Nasopharyngeal Carcinoma/metabolism , Pyrimidinones/pharmacology , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/metabolism , Mice , MicroRNAs/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , RNA, Messenger/metabolism , RNA, Small Interfering , Wnt Signaling Pathway/drug effects
12.
J Mol Neurosci ; 34(1): 63-75, 2008.
Article in English | MEDLINE | ID: mdl-18157659

ABSTRACT

Telomerase is an enzyme composed of a catalytic subunit (TERT) and RNA template (TR), which specifically elongates telomeres and prevents cellular senescence. Although telomerase cannot be detected in most human somatic tissues, including the nervous system, it can be detected in teleost tissues. To facilitate the investigation of telomerase function in the teleost visual system, the coding sequence of zebrafish TERT is revealed and cloned. Immunoblot, immunohistochemistry, reverse transcription polymerase chain reaction (RT-PCR), and telomeric repeats amplification protocol (TRAP) assay are used to assess the expression of telomerase at mRNA, protein, and functional levels in zebrafish retina. Based on the amino acid sequence of mouse TERT, a full-length telomerase reverse transcriptase cDNA of zebrafish has been isolated and cloned. The deduced protein sequence contains 1,091 amino acid residues and a predicted molecular mass of 126 kDa. Multiple alignment shows that the protein sequence contains the conserved motifs and residues found in TERT of other species. RT-PCR and TRAP assay has detected TERT mRNA expression and telomerase activity, respectively, in all tissues examined, including the retina and the brain. The presence of telomerase activity indicates that a fully functional form of telomerase can be found in the retina. Immunohistochemistry reveals that most neurons in zebrafish retina express TERT in the cell nucleus. The presence of telomerase in different tissues may be associated with the indeterminate growth of teleost. However, teleost retinal neurons are post-mitotic and do not further divide under normal situation. The expression of telomerase activity and TERT in retina implies that telomerase has functions other than the elongation of telomere. These findings could provide new insights on telomerase function in the nervous system.


Subject(s)
Telomerase/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Brain/cytology , Brain/metabolism , Cell Differentiation/genetics , Cloning, Molecular , Conserved Sequence , Molecular Sequence Data , Molecular Weight , Neurons/metabolism , Phylogeny , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Retina/metabolism , Sequence Homology, Amino Acid , Telomerase/genetics , Telomerase/isolation & purification , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/isolation & purification
14.
Oncol Rep ; 13(6): 1029-32, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15870917

ABSTRACT

Lung cancer is one of the most common causes of cancer death worldwide. Although many efforts have been made to explore the mechanisms involved in the development of lung cancer, the genetic events involved in the pathogenesis of lung cancer are still unclear. For a better mechanistic scope of study, a well-established cellular model is essential. We report the establishment of a squamous cell carcinoma (SCC) cell line of human lung, SCC-37. Chromosomal abnormalities and global genomic alterations of SCC-37 were studied by spectral karyotyping (SKY) and comparative genomic hybridization (CGH), respectively. Results showed that SCC-37 was a hypodiploid with complex chromosomal rearrangements. Some of the alterations, such as the gain of 1q25-qter in SCC-37, have been correlated to the tumor recurrence of non-small cell lung cancer (NSCLC). Other interesting findings include the amplification of 3q25-qter and 12q13, suggesting the existence of important oncogenes in the amplicons. This cell line may thus provide a useful cellular resource for studying the pathogenesis of SCC of the lung in the future.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Aged , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 3/genetics , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Male , Nucleic Acid Hybridization , Spectral Karyotyping , Telomerase/metabolism , Tumor Cells, Cultured
15.
World J Gastroenterol ; 11(14): 2055-60, 2005 Apr 14.
Article in English | MEDLINE | ID: mdl-15810068

ABSTRACT

AIM: Microsatellites are the repeated DNA sequences scattered widely within the genomes and closely linked with many important genes. This study was designed to characterize the changes of microsatellite DNA loss of heterozygosity (LOH) in esophageal carcinogenesis. METHODS: Allelic deletions in 32 cases of matched precancerous, cancerous and normal tissues were examined by syringe microdissection under an anatomic microscope and microsatellite polymorphism analysis using 15 polymorphic markers on chromosomes 3p, 5q, 6p, 9p, 13q, 17p, 17q and 18q. RESULTS: Microsatellite DNA LOH was observed in precancerous and cancerous tissues, except D9S1752. The rate of LOH increased remarkably with the lesions progressed from basal cell hyperplasia (BCH) to squamous cell carcinoma (SCC) (P<0.05). Three markers, D9S171, D13S260 and TP53, showed the highest incidence of LOH (>60%). LOH loci were different in precancerous and cancerous tissues. LOH in D3S1234 and TP53 was the common event in different lesions from the same patients. CONCLUSION: Microsatellite DNA LOH occurs in early stage of human esophageal carcinogenesis, even in BCH. With the lesion progressed, gene instability increases, the accumulation of this change may be one of the important mechanisms driving precancerous lesions to cancer.


Subject(s)
Carcinoma, Basal Cell/genetics , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Loss of Heterozygosity , Adult , Aged , Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/pathology , China/epidemiology , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/pathology , Female , Humans , Incidence , Male , Microsatellite Repeats , Middle Aged , Neoplasm Staging
16.
Sci Rep ; 5: 9979, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25897700

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an EBV-associated epithelial malignancy prevalent in southern China. Presence of treatment-resistant cancer stem cells (CSC) may associate with tumor relapse and metastasis in NPC. ICG-001 is a specific CBP/ß-catenin antagonist that can block CBP/ß-catenin-mediated transcription of stem cell associated genes and enhance p300/ß-catenin-mediated transcription, thereby reducing the CSC-like population via forced differentiation. In this study, we aimed to evaluate the effect of ICG-001 on the CSC-like population, and the combination effect of ICG-001 with cisplatin in the C666-1 EBV-positive NPC cells. Results showed that ICG-001 inhibited C666-1 cell growth and reduced expression of CSC-associated proteins with altered expression of epithelial-mesenchymal transition (EMT) markers. ICG-001 also inhibited C666-1 tumor sphere formation, accompanied with reduced SOX2(hi)/CD44(hi) CSC-like population. ICG-001 was also found to restore the expression of a tumor suppressive microRNA-145 (miR-145). Ectopic expression of miR-145 effectively repressed SOX2 protein expression and inhibited tumor sphere formation. Combination of ICG-001 with cisplatin synergistically suppressed in vitro growth of C666-1 cells and significantly suppressed growth of NPC xenografts. These results suggested that therapeutically targeting of the CBP/ß-catenin signaling pathway with ICG-001 can effectively reduce the CSC-like population and combination with cisplatin can effectively suppress the growth of NPC.


Subject(s)
Antineoplastic Agents/toxicity , Cell Proliferation/drug effects , Cisplatin/toxicity , Signal Transduction/drug effects , beta Catenin/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/toxicity , Carcinoma , Cell Line, Tumor , Cisplatin/therapeutic use , Drug Synergism , Epithelial-Mesenchymal Transition/drug effects , Herpesvirus 4, Human/isolation & purification , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , Microscopy, Confocal , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pyrimidinones/therapeutic use , Pyrimidinones/toxicity , RNA Interference , RNA, Small Interfering/metabolism , SOXB1 Transcription Factors/antagonists & inhibitors , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transplantation, Heterologous , p300-CBP Transcription Factors/antagonists & inhibitors
17.
FEBS Open Bio ; 4: 128-40, 2014.
Article in English | MEDLINE | ID: mdl-24490137

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a prevalent malignancy in Southeast Asia among the Chinese population. Aberrant regulation of transcripts has been implicated in many types of cancers including NPC. Herein, we characterized mRNA and miRNA transcriptomes by RNA sequencing (RNASeq) of NPC model systems. Matched total mRNA and small RNA of undifferentiated Epstein-Barr virus (EBV)-positive NPC xenograft X666 and its derived cell line C666, well-differentiated NPC cell line HK1, and the immortalized nasopharyngeal epithelial cell line NP460 were sequenced by Solexa technology. We found 2812 genes and 149 miRNAs (human and EBV) to be differentially expressed in NP460, HK1, C666 and X666 with RNASeq; 533 miRNA-mRNA target pairs were inversely regulated in the three NPC cell lines compared to NP460. Integrated mRNA/miRNA expression profiling and pathway analysis show extracellular matrix organization, Beta-1 integrin cell surface interactions, and the PI3K/AKT, EGFR, ErbB, and Wnt pathways were potentially deregulated in NPC. Real-time quantitative PCR was performed on selected mRNA/miRNAs in order to validate their expression. Transcript sequence variants such as short insertions and deletions (INDEL), single nucleotide variant (SNV), and isomiRs were characterized in the NPC model systems. A novel TP53 transcript variant was identified in NP460, HK1, and C666. Detection of three previously reported novel EBV-encoded BART miRNAs and their isomiRs were also observed. Meta-analysis of a model system to a clinical system aids the choice of different cell lines in NPC studies. This comprehensive characterization of mRNA and miRNA transcriptomes in NPC cell lines and the xenograft provides insights on miRNA regulation of mRNA and valuable resources on transcript variation and regulation in NPC, which are potentially useful for mechanistic and preclinical studies.

19.
Cancer Cell ; 26(4): 565-76, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25220446

ABSTRACT

Epstein-Barr virus-induced lymphoproliferative disease (EBV-LPD) after transplantation remains a serious and life-threatening complication. Herein we showed that the aminobisphosphonate pamidronate-expanded human Vγ9Vδ2-T cells efficiently killed EBV-transformed autologous lymphoblastoid B cell lines (EBV-LCL) through γ/δ-TCR and NKG2D receptor triggering and Fas and TRAIL engagement. By inoculation of EBV-LCL in Rag2(-/-)γc(-/-) mice and humanized mice, we established lethal EBV-LPD with characteristics close to those of the human disease. Adoptive transfer of pamidronate-expanded Vγ9Vδ2-T cells alone effectively prevented EBV-LPD in Rag2(-/-)γc(-/-) mice and induced EBV-LPD regression in EBV(+) tumor-bearing Rag2(-/-)γc(-/-) mice. Pamidronate treatment inhibited EBV-LPD development in humanized mice through selective activation and expansion of Vγ9Vδ2-T cells. This study provides proof-of-principle for a therapeutic approach using pamidronate to control EBV-LPD through Vγ9Vδ2-T cell targeting.


Subject(s)
B-Lymphocytes/immunology , Herpesvirus 4, Human/physiology , Lymphocyte Activation , Lymphoproliferative Disorders/physiopathology , T-Lymphocytes/immunology , Animals , Humans , Mice
20.
Lab Invest ; 83(5): 697-709, 2003 May.
Article in English | MEDLINE | ID: mdl-12746479

ABSTRACT

Undifferentiated nasopharyngeal carcinoma (NPC) is closely associated with EBV infection, and the EBV-encoded latent membrane protein 1 (LMP1) is frequently detected in NPC. However, little is known about the pathologic roles of LMP1 in this disease. Recently, we reported the morphologic transformation and increased expression of the LAMC2 and ITGalpha6 genes in LMP1-expressing NPC cell lines. In this study, we further examine the effects of LMP1 in an immortalized nasopharyngeal epithelial cell line called NP69. This cell line was established from primary nonmalignant nasopharyngeal epithelial cells and may represent a model of premalignant nasopharyngeal epithelial cells. LMP1 induced many phenotypic changes in NP69 cells. These include morphologic transformation, increased cell proliferation, anchorage-independent growth, resistance to serum free-induced cell death, and enhanced cell migration and invasion. In addition, expression array analysis identified 28 genes that demonstrated a more than 2-fold difference in expression of NP69 cells expressing LMP1 when compared with a vector control. Two of the up-regulated genes (VEGF and vimentin) identified have been previously reported as LMP1 targets. The majority of the identified genes are associated with cell growth, differentiation, cell shape, and invasion. The present findings support the proposed roles of LMP1 in promoting cell transformation, migration, and invasion in premalignant nasopharyngeal epithelial cells. The present study also indicates the activation of the Ras/MAPK pathway in LMP1-expressing cells, which may be involved in mediating some of the transforming effects of LMP1 observed in nasopharyngeal epithelial cells.


Subject(s)
Nasopharyngeal Neoplasms/etiology , Nasopharynx/pathology , Viral Matrix Proteins/physiology , Blotting, Western , Cell Differentiation , Cell Division , Cell Line , Cell Movement , Cell Survival , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Profiling , Humans , Mitogen-Activated Protein Kinases/metabolism , Nasopharynx/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL