Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Invest New Drugs ; 41(5): 699-709, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37572231

ABSTRACT

Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC) is endemic to parts of Asia and overexpression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α are common in NPC. Anti-vascular agents have known clinical activity in patients with recurrent/ metastatic NPC and in this study, we investigated the anti-tumor effect of BI 836880, a humanized bispecific nanobody against VEGF and angiopoietin-2 (Ang2), in preclinical models of EBV-positive and EBV-negative NPC. The efficacy of BI 836880 was also compared with bevacizumab, a recombinant humanized monoclonal antibody against VEGF. We found that BI 836880 could exert growth-inhibitory effect on endothelial cells (HUVEC-C) and the EBV-negative NPC cell line (HK1), but to a lesser extent in the EBV-positive NPC cell lines, C17C and C666-1. In patients-derived xenograft (PDX) models of NPC - Xeno-2117 and Xeno-666, BI 836880 could suppress tumor growth and Ki67, as well as induce tumor necrosis and reduce microvessel density. Moreover, treatment with BI 836880 increased the level of macrophage infiltration in both PDX tumor models of NPC, suggesting that BI 836880 may exert immunomodulatory effect on the NPC immune microenvironment. When compared with bevacizumab, BI 836880 appeared to show at least comparable activity as bevacizumab in terms of its anti-proliferative and anti-angiogenic effects. This study showed that BI 836880 has anti-proliferative, anti-angiogenic and possibly immunomodulatory effect in clinical models of NPC, therefore the dual targeting of VEGF and Ang2 signaling in NPC should be further investigated.

2.
Cell Death Differ ; 24(6): 1133, 2017 06.
Article in English | MEDLINE | ID: mdl-27636105

ABSTRACT

Retraction to: Cell Death Differ 2016;23(9):1471­1482. doi:10.1038/cdd.2016.32

3.
Cell Death Differ ; 23(9): 1471-82, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27058318

ABSTRACT

Radioresistance is a major obstacle in successful clinical cancer radiotherapy, and the underlying mechanisms are not clear. Here we show that IKKα-mediated miR-196a biogenesis via interaction with Drosha regulates the sensitivity of nasopharyngeal carcinoma (NPC) cells to radiotherapy. Phosphorylation of IKKα at T23 site (p-IKKαT23) promotes the binding of IKKα to Drosha that accelerates the processing of miR-196a primary transcripts, leading to increased expressions of both precursor and mature miR-196a. Dephosphorylation of p-IKKαT23 downregulates miR-196a expression and promotes the resistance of NPC cells to radiation treatment. The miR-196a mimic suppresses while its inhibitor promotes the resistance of NPC to radiation treatment. Importantly, the expression of p-IKKαT23 is positively related to the expression of miR-196a in human NPC tissues, and expression of p-IKKαT23 and miR-196a is inversely correlated with NPC clinical radioresistance. Thus, our studies establish a novel mechanistic link between the inactivation of IKKαT23-Drosha-miR-196a pathway and NPC radioresistance, and de-inactivation of IKKαT23-Drosha-miR-196a pathway would be an efficient way to restore the sensitivity of radioresistant NPC to radiotherapy.

4.
Invest New Drugs ; 33(4): 985-91, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26084990

ABSTRACT

BACKGROUND: This study investigated the activity of MK-2206, an AKT inhibitor, in metastatic or recurrent nasopharyngeal carcinoma (NPC). METHOD: Oral MK-2206 at a dose of 200 mg was administered on days 1, 8, 15 and 22 of a 28-day cycle until progression. Plasma EBV DNA clearance during the first month of treatment was measured, and archived tumors were analyzed for the expression of AKT and PIK3CA mutation and PIK3CA amplification. The dual primary endpoint was objective response rate and 6-month progression-free survival (PFS) rate. RESULTS: 21 patients were enrolled and one patient achieved a partial response (5 %) and 11 had stable disease (52 %), with a median PFS of 3.5 months (95 % confidence interval, CI: 0.9-7.3). The 6-month PFS rate was 43 % (95 % CI: 22-66 %) and the median OS was 10 months (95 % CI: 5.9 months-not reached). Seven patients (33 %) experienced grade 3 toxicities which could be related to MK-2206. Macular-papular rash was the most common (n = 6), followed by hyperglycemia (n = 2) and fatigue (n = 1). In the 12 tumor samples analyzed, PIK3CA amplification was detected in one patient's primary NPC, who had SD lasting over 12 months. Patients with decreasing EBV DNA values over time were more likely to be alive and progression-free for at least 6 months than those without a decrease (p = 0.001). CONCLUSION: The study was terminated due to the limited activity observed in this heavily pre-treated group of patients. Further studies are needed to elucidate the optimal way of selecting patients for AKT inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Heterocyclic Compounds, 3-Ring/therapeutic use , Nasopharyngeal Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Carcinoma , DNA, Viral/blood , Disease-Free Survival , Female , Herpesvirus 4, Human/genetics , Heterocyclic Compounds, 3-Ring/adverse effects , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/blood , Nasopharyngeal Neoplasms/virology , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/virology , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Treatment Outcome
5.
Pediatr Surg Int ; 30(1): 107-11, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24085513

ABSTRACT

AIM OF THE STUDY: Anatomical variations on venous drainage in varicoceles are under-reported. We report our experience in scrotal antegrade sclerotherapy (SAS) for adolescent varicoceles. METHODS: Since 2011, 15 consecutive boys with left varicoceles were recruited. Under general anaesthesia, a 5-mm transverse incision was made at scrotal neck, testicular vein was cannulated at pampiniform plexus with venogram performed. Foam sclerosant by mixing sodium tetradecyl sulphate (STS), Lipiodol(®) and air was slowly injected under fluoroscopy. Postoperatively the patients were followed-up for varicocele grading, testicular size, and complications. MAIN RESULTS: Median age at operation was 14 (10-19) years. 80 % had grade three varicoceles, 33.3 % had smaller left testis before operation. Intra-operative venogram showed three different anatomical variations. Group I: eleven patients (73.3 %) had single distinct internal spermatic vein; Group II: two patients demonstrated duplication of internal spermatic vein draining into left renal vein; Group III: two patients had pampiniform plexus draining to iliac and/or paraspinal veins. SAS was performed in Group I and II patients. Sclerosant volume injected ranged from 1.5 to 4.5 ml. In Group III patients, surgical ligation of testicular veins was performed rather than SAS to avoid uncontrolled systemic sclerosant spillage. Mean length of stay was 1.13 day. One patient with scrotal haematoma and one other with minor wound dehiscence were managed conservatively. Mean follow-up period was 10.9 (1-22) months. Thirteen patients (86.7 %) achieved varicocele grading ≤ 1. There was no postoperative testicular atrophy, hydrocele and epididymo-orchitis. CONCLUSION: Scrotal antegrade sclerotherapy using STS foam is a safe and effective treatment for adolescent varicoceles. Anatomical variations on venous drainage in varicoceles are common.


Subject(s)
Sclerotherapy/methods , Scrotum/blood supply , Varicocele/therapy , Adolescent , Adult , Child , Follow-Up Studies , Humans , Male , Sclerosing Solutions/therapeutic use , Scrotum/anatomy & histology , Sodium Tetradecyl Sulfate/therapeutic use , Testis/anatomy & histology , Testis/blood supply , Treatment Outcome , Young Adult
6.
Int J Cancer ; 130(5): 1036-45, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21400511

ABSTRACT

MicroRNAs (miRNAs) regulate mRNA stability and protein expression, and certain miRNAs have been demonstrated to act either as oncogenes or tumor suppressors. Differential miRNA expression signatures have been documented in many human cancers but the role of miRNAs in endometrioid endometrial cancer (EEC) remains poorly understood. This study identifies significantly dysregulated miRNAs of EEC cells, and characterizes their impact on the malignant phenotype. We studied the expression of 365 human miRNAs using Taqman low density arrays in EECs and normal endometriums. Candidate differentially expressed miRNAs were validated by quantitative real-time PCR. Expression of highly dysregulated miRNAs was examined in vitro through the effect of anti-/pre-miRNA transfection on the malignant phenotype. We identified 16 significantly dysregulated miRNAs in EEC and 7 of these are novel findings with respect to EEC. Antagonizing the function of miR-7, miR-194 and miR-449b, or overexpressing miR-204, repressed migration, invasion and extracellular matrix-adhesion in HEC1A endometrial cancer cells. FOXC1 was determined as a target gene of miR-204, and two binding sites in the 3'-untranslated region were validated by dual luciferase reporter assay. FOXC1 expression was inversely related to miR-204 expression in EEC. Functional analysis revealed the involvement of FOXC1 in migration and invasion of HEC1A cells. Our results present dysfunctional miRNAs in endometrial cancer and identify a crucial role for miR-204-FOXC1 interaction in endometrial cancer progression. This miRNA signature offers a potential biomarker for predicting EEC outcomes, and targeting of these cancer progression- and metastasis-related miRNAs offers a novel potential therapeutic strategy for the disease.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasm Invasiveness , 3' Untranslated Regions , Cell Adhesion , Cell Line, Tumor , Cell Movement , Endometrial Neoplasms , Endometrium/metabolism , Female , Gene Expression Profiling , Humans , Transfection , Validation Studies as Topic
7.
Mol Carcinog ; 51(12): 963-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22006341

ABSTRACT

2-methoxyestradiol (2ME2), an endogenous metabolite of 17-ß-estradiol, has been shown to induce apoptosis and cell cycle arrest in various tumor models. We have previously shown that 2ME2 induced endoreduplication in a well-differentiated nasopharyngeal carcinoma (NPC) HK-1 and a poorly differentiated C666-1 cell line. In the present study, we studied the survival factors involved in 2ME2-induced endoreduplicating NPC cells. In the HK-1 cells, knockdown of BcL-xL expression by siRNA resulted in the reduction of endoreduplication and an increase in the percentage of apoptosis. Further mechanistic study revealed that 2ME2 enhanced the expression of the phosphorylated form of STAT5 (p-STAT5-Y694), but not p-STAT3 (Y705) and p-STAT3 (S727), in the nucleus of HK-1 cells. Pre-treatment of cells with JAK/STAT inhibitor AG490 and STAT5 inhibitor resulted not only in the reduced expression of Bcl-xL, but also reduced the percentage of endoreduplicating cells. In contrast, 2ME2 enhanced the expression of p-STAT3 in the poorly differentiated C666-1 cells. Pharmacological inhibition of STAT3 or Bcl-2/xL resulted in a decrease in endoreduplication of C666-1 cells. Taken together, the expression of p-STAT5 and p-STAT3 was upregulated in 2ME2-induced endoreduplicating HK-1 and C666-1 cells, respectively. Combination of 2ME2 with Bcl-2/xL inhibitor is a novel strategy to reduce the formation of endoreduplicating cells during chemotherapeutic treatment of NPC. © 2011 Wiley Periodicals, Inc.


Subject(s)
Endoreduplication/drug effects , Estradiol/analogs & derivatives , Nasopharyngeal Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , STAT3 Transcription Factor/physiology , STAT5 Transcription Factor/physiology , bcl-X Protein/physiology , 2-Methoxyestradiol , Base Sequence , Blotting, Western , Carcinoma , Cell Line, Tumor , Estradiol/pharmacology , Flow Cytometry , Humans , Nasopharyngeal Carcinoma , RNA, Small Interfering
8.
Gene Ther ; 19(8): 844-51, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21918552

ABSTRACT

Antiangiogenesis is an appealing anticancer approach but requires continued presence of the antiangiogenic agents, which can be remedied by gene therapy. Baculovirus is an emerging gene delivery vector but only mediates transient expression (<7 days); thus, this study primarily aimed to develop a hybrid baculovirus for sustained antiangiogenic gene expression and cancer therapy. We first constructed plasmids featuring adeno-associated virus inverted terminal repeats (AAV ITRs), oriP/Epstein-Barr virus-expressed nuclear antigen 1 (EBNA1) or Sleeping Beauty (SB) transposon and compared their efficacies in terms of persistent expression. In human embryonic kidney (HEK293) cells, AAV ITR failed to prolong the expression while oriP/EBNA1 moderately extended the expression to 35 days. In contrast, the SB system led to stable expression beyond 77 days even without antibiotic selection. Given this finding, we constructed a hybrid SB baculovirus expressing the SB transposase and harboring the transgene cassette flanked by inverted repeat/direct-repeat (IR/DR) elements recognizable by SB. The hybrid SB baculovirus efficiently transduced mammalian cells and mediated an expression duration longer than that by conventional baculoviruses, thanks to the transgene persistence and integration. The SB baculovirus (Bac-SB-T2hEA/w) expressing the antiangiogenic fusion protein comprising endostatin and angiostatin (hEA) also enabled prolonged hEA expression. With sustained hEA expression, Bac-SB-T2hEA/w repressed the angiogenesis in vivo, hindered the growth of two different tumors (prostate tumor allografts and human ovarian tumor xenografts) in mice and extended the life span of animals. These data altogether implicated the potential of the hybrid SB-baculovirus vector for prolonged hEA expression and for the treatment of multiple types of angiogenesis-dependent tumors.


Subject(s)
Baculoviridae/genetics , Genetic Therapy , Genetic Vectors , Animals , Dependovirus/genetics , Female , Gene Expression , HEK293 Cells , Humans , Male , Mice , Ovarian Neoplasms/therapy , Prostatic Neoplasms/therapy , Recombination, Genetic , Terminal Repeat Sequences , Transduction, Genetic , Transgenes , Transposases/genetics , Xenograft Model Antitumor Assays
9.
Oncogene ; 31(15): 1975-87, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-21874044

ABSTRACT

RASSF1A is a key tumor-suppressor gene that is often inactivated in a wide variety of solid tumors. Studies have illustrated that RASSF1A plays vital roles in the regulation of cell-cycle progression and functions as a guardian of mitosis. Nevertheless, the precise mechanism of RASSF1A-dependent regulation of mitosis remains largely unclear. APC/C(Cdc20) is the master switch and regulator of mitosis. The activity of APC/C(Cdc20) is tightly controlled by phosphorylation and specific inhibitors to ensure the sequential ubiquitination of downstream targets. Here, we report on the novel finding of a regulated circuitry that controls the timely expression and hence activity of APC/C(Cdc20) during mitosis. Our study showed that RASSF1A and APC/C(Cdc20) form a molecular relay that regulates the APC/C(Cdc20) activity at early mitosis. We found that RASSF1A inhibits APC/C(Cdc20) function through its D-box motifs. Paradoxically, RASSF1A was also demonstrated to be ubiquitinated by APC/C(Cdc20) in vitro and degraded at prometaphase despite of active spindle checkpoint presence. The first two unique D-boxes at the N-terminal of RASSF1A served as specific degron recognized by APC/C(Cdc20). Importantly, we found that Aurora A and Aurora B directly phosphorylate RASSF1A, a critical step by which RASSF1A switches from being an inhibitor to a substrate of APC/C(Cdc20) during the course of mitotic progression. As a result of RASSF1A degradation, APC/C(Cdc20) can then partially activate the ubiquitination of Cyclin A in the presence of spindle checkpoint. This circuitry is essential for the timely degradation of Cyclin A. To conclude, our results propose a new model for RASSF1A-APC/C(Cdc20) interaction in ensuring the sequential progression of mitosis.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Cycle Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Aurora Kinase B , Aurora Kinases , Cdc20 Proteins , Cyclin A/metabolism , HEK293 Cells , HeLa Cells , Humans , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Transfection , Ubiquitination
10.
Oncogene ; 30(9): 1127-34, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21057531

ABSTRACT

c-Met represents an important emerging therapeutic target in cancer. In this study, we demonstrate the mechanism by which c-Met tyrosine kinase inhibition inhibits tumor growth in a highly invasive Asian-prevalent head and neck cancer, nasopharyngeal cancer (NPC). c-Met tyrosine kinase inhibitors (TKIs; AM7 and c-Met TKI tool compound SU11274) downregulated c-Met phosphorylation, resulting in marked inhibition of NPC cell growth and invasion. Strikingly, inhibition of c-Met resulted in significant downregulation of TP53-induced Glycolysis and Apoptosis Regulator (TIGAR) and subsequent depletion of intracellular NADPH. Importantly, overexpression of TIGAR ameliorated the effects of c-Met kinase inhibition, confirming the importance of TIGAR downregulation in the growth inhibitory activity of c-Met TKI. The effects of c-Met inhibition on TIGAR and NADPH levels were observed with two different c-Met TKIs (AM7 and SU11274) and with multiple cell lines. As NADPH provides a crucial reducing power required for cell survival and proliferation, our findings reveal a novel mechanistic action of c-Met TKI, which may represent a key effect of c-Met kinase inhibition. Our data provide the first evidence linking c-Met, TIGAR and NADPH regulation in human cancer cells suggesting that inhibition of a tyrosine kinase/TIGAR/NADPH cascade may have therapeutic applicability in human cancers.


Subject(s)
Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , NADP/biosynthesis , Nasopharyngeal Neoplasms/metabolism , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrimidinones/pharmacology , Quinolines/pharmacology , Sulfonamides/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Down-Regulation , Humans , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Phosphoric Monoester Hydrolases , Phosphorylation , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
11.
Oncogene ; 30(13): 1518-30, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21119603

ABSTRACT

The fibroblast growth factor 8b (FGF8b) oncogene is known to be primarily involved in the tumorigenesis and progression of hormone-related cancers. Its role in other epithelial cancers has not been investigated, except for esophageal cancer, in which FGF8b overexpression was mainly found in tumor biopsies of male patients. These observations were consistent with previous findings in these cancer types that the male sex-hormone androgen is responsible for FGF8b expression. Nasopharyngeal carcinoma (NPC) is a highly metastatic cancer of head and neck commonly found in Asia. It is etiologically associated with Epstein-Barr Virus (EBV) infection, inflammatory tumor microenvironment and relatively higher male predominance. Here, we reported for the first time that FGF8b is overexpressed in this EBV-associated non-hormone-related cancer of the head and neck, NPC. More importantly, overexpression of FGF8b mRNA and protein was detected in a large majority of NPC tumors from both male and female genders, in addition to multiple NPC cell lines. We hypothesized that FGF8b overexpression may contribute to NPC tumorigenesis. Using EBV-associated NPC cell lines, we demonstrated that specific knockdown of FGF8b by small interfering RNA inhibited cell proliferation, migration and invasion, whereas exogenous FGF8b stimulated these multiple phenotypes. Further mechanistic investigation revealed that in addition to NF-κB signaling (a major inflammatory signaling pathway known to be activated in NPC), an important EBV oncoprotein, the latent membrane protein 1 (LMP1), was found to be a direct inducer of FGF8b overexpression in NPC cells, whereas androgen (testosterone) has minimal effect on FGF8b expression in EBV-associated NPC cells. In summary, our study has identified LMP1 as the first viral oncogene capable of directly inducing FGF8b (an important cellular oncogene) expression in human cancer cells. This novel mechanism of viral-mediated FGF8 upregulation may implicate a new role of oncoviruses in human carcinogenesis.


Subject(s)
Fibroblast Growth Factor 8/physiology , Gene Expression Regulation, Neoplastic , Herpesvirus 4, Human/pathogenicity , Oncogenes , Carcinoma , Cell Movement , Cell Proliferation , Female , Fibroblast Growth Factor 8/antagonists & inhibitors , Fibroblast Growth Factor 8/genetics , Humans , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/physiology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplasm Invasiveness , RNA, Messenger/analysis , RNA, Small Interfering/genetics , Viral Matrix Proteins/physiology
12.
Biochem Pharmacol ; 79(6): 825-41, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19883629

ABSTRACT

2-Methoxyestradiol (2ME2) is a normal physiological metabolite of 17beta-estradiol with anti-proliferative and anti-angiogenic activities. The purpose of this study is to elucidate the mechanism whereby 2ME2 induces endoreduplication of the well-differentiated nasopharyngeal carcinoma (NPC) cells. We report here that 2ME2 induces G2/M phase cell cycle arrest followed by endoreduplication of the well-differentiated HK-1 cells. The increase in chromosome number was confirmed by cytogenetic study. Analysis of stress signaling pathways revealed the phosphorylation activation of ERK, JNK and p38 MAPKs at various times after 2ME2 treatment. Pre-treatment of 2ME2-treated HK-1 cells with JNK inhibitor (SP600125), ERK inhibitor (PD98059) and p38 MAPK inhibitor (SB203580) resulted in the reduction of endoreduplicating cells. Furthermore, the increase in the phosphorylation of JNK was accompanied by an increase in the reactive oxygen species. In addition, endoreduplication was observed in cells after treatment with superoxide donor, 2,3-dimethoxy-1,4-naphoquinone (DMNQ). Confocal microscopic analysis also revealed the increase in mitochondrial superoxide anion in 2ME2-treated HK-1 cells. Pre-treatment of HK-1 cells with superoxide dismutase mimetic 2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPO) or overexpressing the mitochondrial enzyme MnSOD resulted in the reduction of phosphorylation of JNK and the formation of endoreduplicating cells. Furthermore, the tubulin filaments in cytoplasm remain intact in 2ME2-treated HK-1 cells after pre-treatment of TEMPO. Our results suggest that 2ME2 induces endoreduplication through the induction of oxidative stress and the activation of MAPK signal pathways. The biological significance of drug-induced endoreduplication will also be discussed.


Subject(s)
Estradiol/analogs & derivatives , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Tubulin Modulators/pharmacology , 2-Methoxyestradiol , Cell Line , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme Activation , Estradiol/pharmacology , Humans , Mitochondria/metabolism
13.
Cell Death Differ ; 16(11): 1469-79, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19575017

ABSTRACT

Overexpression of BMI1 correlates with cancer development, progression, and therapy failure; however, the underlying molecular mechanisms remain to be fully elucidated. Using the C666-1 nasopharyngeal cancer (NPC) model, the role of BMI1 in mediating response of NPC cells to radiation therapy (RT) was investigated. The results showed a novel radioresistance function for BMI1 in NPC, wherein BMI1 depletion sensitized NPC cells to RT. Cell cycle analysis and transmission electron microscopy (TEM) showed apoptosis as the major mode of cell death, and the mitochondria as a primary targeted cellular organelle. Genome-wide microarray and pathway analyses revealed that the P53 pathway is a critical mediator of this process. Cotransfection with siP53 rescued C666-1 cells from cytotoxicity upon BMI1 depletion and RT, thereby corroborating the role for P53. Pretreatment with the antioxidant, Trolox, inhibited apoptosis, indicating that production of reactive oxygen species (ROS) is also mediating cytotoxicity. In vivo, BMI1 depletion combined with RT abrogated tumor-forming capacity in SCID mice, showing the relevance of this process in a more complex tumor environment. Hence, we show a novel role for BMI1 in conferring radioresistance in cancer cells through the downregulation of p53-mediated apoptosis. These results suggest a potential strategy of BMI1 depletion combined with RT for tumors wherein BMI1 appears to be driving disease progression.


Subject(s)
Apoptosis , Nasopharyngeal Neoplasms/radiotherapy , Nuclear Proteins/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Chromans/pharmacology , Disease Progression , Gene Expression Profiling , Gene Knockdown Techniques , Histones/metabolism , Humans , Mice , Microarray Analysis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
14.
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
15.
J Exp Clin Cancer Res ; 26(1): 77-81, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17550135

ABSTRACT

Human Papillomavirus (HPV) and Hepatitis B virus (HBV) are known aetiology of cervical and hepatocellular carcinoma, respectively. Both diseases share a similar clinical course, that is, the vast majority of those infected by these two viruses can eradicate the viruses spontaneously. A small sub-group who fails to clear the virus becomes chronic carrier and can progress to carcinoma many years later. We postulated that patients with pre-malignant or malignant cervical lesion are at increased risk of becoming chronic HBV carrier if infected, which may be attributed to inherent immunological deficiency against viral infection. We tested HBV carrier status from 288 patients with cervical carcinoma, 242 patients with high grade cervical intra-epithelial neoplasia (CIN) and 311 women with neither of the above conditions as control subjects. The HBV carrier rate in the Cancer Group, CIN Group and Control Group was 21.4%, 24.1% and 10.6%. The carrier rate was significantly higher in both the Cancer Group (p<0.01) and the CIN Group (p<0.01), compared to the Control Group. Our study suggests that a common immunological mechanism is involved in eradication of HBV and HPV infections and inherent immuno-deficiency might lead to an association of HBV carrier status with cervical carcinoma. Further studies are needed to confirm our findings and delineate the mechanism involved.


Subject(s)
Carrier State , Hepatitis B, Chronic/transmission , Papillomavirus Infections/complications , Uterine Cervical Dysplasia/complications , Uterine Cervical Neoplasms/complications , Adult , Aged , Case-Control Studies , Female , Hepatitis B Antibodies/blood , Hepatitis B Core Antigens/blood , Hepatitis B, Chronic/epidemiology , Hepatitis B, Chronic/etiology , Hepatitis B, Chronic/immunology , Hong Kong/epidemiology , Humans , Incidence , Middle Aged , Papillomavirus Infections/epidemiology , Risk Assessment , Risk Factors , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Dysplasia/etiology , Uterine Cervical Dysplasia/immunology , Uterine Cervical Dysplasia/virology
16.
Eur J Gynaecol Oncol ; 28(2): 98-102, 2007.
Article in English | MEDLINE | ID: mdl-17479669

ABSTRACT

OBJECTIVE: To evaluate the role of aortic lymphadenectomy in the management of endometrial carcinoma. METHODS: Clinical notes of 163 patients with endometrial carcinoma were reviewed. All patients had peritoneal cytology, total abdominal hysterectomy, bilateral salpingo-oophorectomy and pelvic lymphadenectomy with or without aortic lymphadenectomy. RESULTS: Seventy-five (46.0%) patients had pelvic lymphadenectomy alone whereas 88 (54.0%) had both pelvic and aortic lymphadenectomy. Thirty-five (21.5%) patients had nodal metastases with positive pelvic and aortic nodes in 26 (16.0%) and 24 (27.3%) patients, respectively. Isolated aortic metastases were found in 17 cases (19.3%). Among 35 patients with nodal metastases, recurrence developed in 15 (42.9%) patients and all except one died within five to 50 months. The remaining patients had a median disease-free period of 55 months (13-93 months). The recurrence rate was higher (63.6%) among patients with upper aortic lymph node metastases, and all those who recurred died of disease within seven to 28 months. CONCLUSIONS: Our data suggest that aortic lymphadenectomy provides both diagnostic and therapeutic value in the management of endometrial carcinoma with high metastatic risk. After surgical removal and adjuvant radiotherapy, patients with nodal metastases achieved a better survival chance.


Subject(s)
Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Lymph Node Excision , Adult , Aged , Aorta, Abdominal , Disease-Free Survival , Female , Follow-Up Studies , Humans , Lymph Nodes/pathology , Lymph Nodes/surgery , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Radiotherapy, Adjuvant , Women's Health
17.
Br J Cancer ; 96(4): 617-22, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17262084

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus-associated disease with high prevalence in Southern Chinese. Using multiparametric flow cytometry, we identified significant expansions of circulating naïve and memory CD4+CD25(high) T cells in 56 NPC patients compared with healthy age- and sex-matched controls. These were regulatory T cells (Treg), as they overexpressed Foxp3 and GITR, and demonstrated enhanced suppressive activities against autologous CD4+CD25- T-cell proliferation in functional studies on five patients. Abundant intraepithelial infiltrations of Treg with very high levels of Foxp3 expression and absence of CCR7 expression were also detected in five primary tumours. Our current study is the first to demonstrate an expansion of functional Treg in the circulation of NPC patients and the presence of infiltrating Treg in the tumour microenvironment. As Treg may play an important role in suppressing antitumour immunity, our findings provide critical insights for clinical management of NPC.


Subject(s)
Forkhead Transcription Factors/biosynthesis , Lymphocytes, Tumor-Infiltrating/immunology , Nasopharyngeal Neoplasms/immunology , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , T-Lymphocytes, Regulatory/immunology , Cell Proliferation , Cells, Cultured , Flow Cytometry/methods , Glucocorticoid-Induced TNFR-Related Protein , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Neoplasm Staging , Sensitivity and Specificity
18.
Oncogene ; 26(21): 3069-80, 2007 May 10.
Article in English | MEDLINE | ID: mdl-17099724

ABSTRACT

Epstein-Barr virus (EBV) infection is closely associated with nasopharyngeal carcinoma (NPC) and can be detected in early premalignant lesions of nasopharyngeal epithelium. The latent membrane protein 1 (LMP1) is an oncoprotein encoded by the EBV and is believed to play a role in transforming premalignant nasopharyngeal epithelial cells into cancer cells. RASSF1A is a tumor-suppressor gene commonly inactivated in many types of human cancer including NPC. In this study, we report a novel function of LMP1, in down-regulating RASSF1A expression in human epithelial cells. Downregulation of RASSF1A expression by LMP1 is dependent on the activation of intracellular signaling of NF-kappaB involving the C-terminal activating regions (CTARs) of LMP1. LMP1 expression also suppresses the transcriptional activity of the RASSF1A core promoter. RASSF1A stabilizes microtubules and regulates mitotic events. Aberrant mitotic spindles and chromosome aberrations are reported phenotypes in RASSF1A inactivated cells. In this study, we observed that LMP1 expression in human epithelial cells could induce aberrant mitotic spindles, disorganized interphase microtubules and aneuploidy. LMP1 expression could also suppress microtubule dynamics as exemplified by tracking movements of the growing tips of microtubules in live cells by transfecting EGFP-tagged EB1 into cells. The aberrant mitotic spindles and interphase microtubule organization induced by LMP1 could be rescued by transfecting RASSF1A expression plasmid into cells. Downregulation of RASSF1A expression by LMP1 may facilitate its role in transformation of premalignant nasopharyngeal epithelial cells into cancer cells.


Subject(s)
Chromosome Aberrations , Down-Regulation/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Microtubules/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Viral Matrix Proteins/physiology , Cell Line , Cell Line, Transformed , Cell Line, Tumor , HeLa Cells , Humans , Microtubules/pathology , NF-kappa B/physiology , Tumor Suppressor Proteins/biosynthesis
19.
Oncogene ; 26(13): 1971-82, 2007 Mar 22.
Article in English | MEDLINE | ID: mdl-17043662

ABSTRACT

Endometrial cancer is the third most common gynecologic malignancy and the ninth most common malignancy for females overall in Hong Kong. Approximately 80% or more of these cancers are endometrioid endometrial adenocarcinomas. The aim of this study was to reveal genes contributing to the development of endometrioid endometrial cancer, which may impact diagnosis, prognosis and treatment of the disease. Whole-genome gene expression analysis was completed for a set of 55 microdissected sporadic endometrioid endometrial adenocarcinomas and 29 microdissected normal endometrium specimens using the Affymetrix Human U133 Plus 2.0 oligonucleotide microarray. Selected genes of interest were validated by quantitative real-time-polymerase chain reaction (qRT-PCR). Pathway analysis was performed to reveal gene interactions involved in endometrial tumorigenesis. Unsupervised hierarchical clustering displayed a distinct separation between the endometrioid adenocarcinomas and normal endometrium samples. Supervised analysis identified 117 highly differentially regulated genes (>or=4.0-fold change), which distinguished the endometrial cancer specimens from normal endometrium. Twelve novel genes including DKK4, ZIC1, KIF1A, SAA2, LOC16378, ALPP2, CCL20, CXCL5, BST2, OLFM1, KLRC1 and MBC45780 were deregulated in the endometrial cancer, and further validated in an independent set of 56 cancer and 29 normal samples using qRT-PCR. In addition, 10 genes were differentially regulated in late-stage cancer, as compared to early-stage disease, and may be involved in tumor progression. Pathway analysis of the expression data from this tumor revealed an interconnected network consisting of 21 aberrantly regulated genes involved in angiogenesis, cell proliferation and chromosomal instability. The results of this study highlight the molecular features of endometrioid endometrial cancer and provide insight into the events underlying the development and progression of endometrioid endometrial cancer.


Subject(s)
Endometrial Neoplasms/metabolism , Gene Expression Profiling , Genome , Signal Transduction , Endometrial Neoplasms/genetics , Female , Hong Kong , Humans , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
20.
Cancer J ; 12(3): 189-93, 2006.
Article in English | MEDLINE | ID: mdl-16803676

ABSTRACT

UNLABELLED: The objective of the present preliminary study was to determine if a difference in the pattern of gene expression exists between tumors that were subsequently found to be sensitive to radiotherapy and tumors found to be resistant to radiotherapy. PATIENTS AND METHODS: A total of 16 patients with invasive squamous cell carcinoma of the uterine cervix were included in this study. All patients were treated with standardized radiotherapy alone. Ten of the tumors were clinically radiosensitive and six were radioresistant. Total RNA, extracted from tumor specimens obtained prior to treatment, was hybridized onto an oligonucleotide microarray with probe sets complementary to over 20,000 transcripts. The genes were first subjected to a statistical filter to identify genes with statistically significant differential expression levels between those that were radiosensitive and those that were radioresistant. A back-propagation neural network was then constructed to model the differences so that patterns could be easily identified. RESULTS: Although a number of genes were found to express differentially between radiosensitive and radioresistant tumors; the 10 most discriminating genes were used to construct the model. Using the expressions from these 10 genes, we found that neural networks constructed from random subsets of the whole data were capable of predicting radiotherapy responses in the remaining subset, which appears stable within the dataset. DISCUSSION: This study shows that such an approach has the potential to differentiate tumor radiosensitivity, although confirmation of such a pattern using other larger independent datasets is necessary before firm conclusions can be drawn.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Radiation Tolerance/genetics , Uterine Cervical Neoplasms/genetics , Adult , Aged , Carcinoma, Squamous Cell/radiotherapy , Female , Gene Expression Profiling , Humans , Middle Aged , Neoplasm Invasiveness/pathology , Oligonucleotide Array Sequence Analysis , RNA, Neoplasm , Tumor Cells, Cultured , Uterine Cervical Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...