Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
J Gen Virol ; 97(12): 3174-3182, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27902314

ABSTRACT

Newcastle disease virus (NDV) is a candidate agent for oncolytic virotherapy. Despite its potential, the exact mechanism of its oncolysis is still not known. Recently, we reported that NDV exhibited an increased oncolytic activity in hypoxic cancer cells. These types of cells negatively affect therapeutic outcome by overexpressing pro-survival genes under the control of the hypoxia-inducible factor (HIF). HIF-1 is a heterodimeric transcriptional factor consisting of a regulated α (HIF-1α) and a constitutive ß subunit (HIF-1ß). To investigate the effects of NDV infection on HIF-1α in cancer cells, the osteosarcoma (Saos-2), breast carcinoma (MCF-7), colon carcinoma (HCT116) and fibrosarcoma (HT1080) cell lines were used in the present study. Data obtained showed that a velogenic NDV infection diminished hypoxia-induced HIF-1α accumulation, leading to a decreased activation of its downstream target gene, carbonic anhydrase 9. This NDV-induced downregulation of HIF-1α occurred post-translationally and was partially abrogated by proteasomal inhibition. The process appeared to be independent of the tumour suppressor protein p53. These data revealed a correlation between NDV infection and HIF-1α downregulation, which highlights NDV as a promising agent to eliminate hypoxic cancer cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/metabolism , Newcastle disease virus/physiology , Oncolytic Viruses/physiology , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neoplasms/genetics , Neoplasms/therapy , Newcastle disease virus/genetics , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Proteolysis , Tumor Suppressor Protein p53/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
2.
Oncol Lett ; 10(4): 2192-2196, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26622817

ABSTRACT

Bortezomib is the first proteasomal inhibitor (PI) to be used therapeutically for treating relapse cases of multiple myeloma and mantle cell lymphoma. A proposed mechanism for its action is that it prevents the proteasomal degradation of proapoptotic proteins, leading to enhanced apoptosis. Although the α subunit of hypoxia-inducible factor (HIF)-1 is not degraded with bortezomib treatment, the heterodimeric HIF-1 fails to transactivate target genes. HIF-1 and HIF-2 are related hypoxia-inducible transcription factors that are important for the survival of hypoxic tumor cells. The majority of reports have focused on the effects of bortezomib on the transcriptional activities of HIF-1, but not HIF-2. The present study investigated the effects of bortezomib on HIF-2 activity in cancer cells with different levels of HIF-1α and HIF-2α subunits. HIF-α subunit levels were detected using specific antibodies, while HIF transcriptional activities were evaluated using immunodetection, reverse transcription-polymerase chain reaction and luciferase reporter assay. Bortezomib treatment was found to suppress the transcription and expression of CA9, a HIF-1-specific target gene; however, it had minimal effects on EPO and GLUT-1, which are target genes of both HIF-1 and HIF-2. These data suggest that bortezomib attenuates the transcriptional activity only of HIF-1, and not HIF-2. This novel finding on the lack of an inhibitory effect of bortezomib on HIF-2 transcriptional activity has implications for the improvement of design and treatment modalities of bortezomib and other PI drugs.

3.
Cell Oncol (Dordr) ; 38(4): 279-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25930675

ABSTRACT

PURPOSE: Newcastle disease virus (NDV) is an oncolytic virus that is known to have a higher preference to cancer cells than to normal cells. It has been proposed that this higher preference may be due to defects in the interferon (IFN) responses of cancer cells. The exact mechanism underlying this process, however, remains to be resolved. In the present study, we examined the antiviral response towards NDV infection of clear cell renal cell carcinoma (ccRCC) cells. ccRCC is associated with mutations of the von Hippel-Lindau tumor suppressor gene VHL, whose protein product is important for eliciting cellular responses to changes in oxygen levels. The most common first line treatment strategy of ccRCC includes IFN. Unfortunately, most ccRCC cases are diagnosed at a late stage and often are resistant to IFN-based therapies. Alternative treatment approaches, including virotherapy using oncolytic viruses, are currently being investigated. The present study was designed to investigate the mechanistic pathways underlying the response of ccRCC cells to oncolytic NDV infection. METHODS AND RESULTS: We found that NDV induces activation of NF-κB in ccRCC cells by inducing phosphorylation and subsequent degradation of IκBα. IκBα was found to be phosphorylated as early as 1 hour post-infection and to result in rapid NF-κB nuclear translocation and activation. Importantly, p38 MAPK phosphorylation was found to occur upstream of the NDV-induced NF-κB activation. Restoration of VHL in ccRCC cells did not result in a reduction of this phosphorylation. A similar phenomenon was also observed in several other cancer-derived cell lines. CONCLUSION: Our data provide evidence for involvement of the p38 MAPK/NF-κB/IκBα pathway in NDV infection and subsequent induction of apoptosis in ccRCC cells.


Subject(s)
I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Newcastle disease virus/growth & development , p38 Mitogen-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Apoptosis/drug effects , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/virology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Host-Pathogen Interactions/drug effects , Humans , Imidazoles/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/virology , MCF-7 Cells , Mutation , NF-KappaB Inhibitor alpha , Newcastle disease virus/physiology , Oncolytic Viruses/physiology , Phosphorylation/drug effects , Pyridines/pharmacology , Signal Transduction/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
4.
J Interferon Cytokine Res ; 33(7): 346-54, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23506478

ABSTRACT

Viral-mediated oncolysis is a promising cancer therapeutic approach offering an increased efficacy with less toxicity than the current therapies. The complexity of solid tumor microenvironments includes regions of hypoxia. In these regions, the transcription factor, hypoxia inducible factor (HIF), is active and regulates expression of many genes that contribute to aggressive malignancy, radio-, and chemo-resistance. To investigate the oncolytic efficacy of a highly virulent (velogenic) Newcastle disease virus (NDV) in the presence or absence of HIF-2α, renal cell carcinoma (RCC) cell lines with defective or reconstituted wild-type (wt) von Hippel-Lindau (VHL) activity were used. We show that these RCC cells responded to NDV by producing only interferon (IFN)-ß, but not IFN-α, and are associated with increased STAT1 phosphorylation. Restoration of wt VHL expression enhanced NDV-induced IFN-ß production, leading to prolonged STAT1 phosphorylation and increased cell death. Hypoxia augmented NDV oncolytic activity regardless of the cells' HIF-2α levels. These results highlight the potential of oncolytic NDV as a potent therapeutic agent in the killing of hypoxic cancer cells.


Subject(s)
Carcinoma, Renal Cell/therapy , Hypoxia/therapy , Interferon-beta/metabolism , Newcastle disease virus , Oncolytic Virotherapy/methods , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/genetics , Humans , Hypoxia/immunology , Phosphorylation/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Transgenes/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics
5.
Cancer Causes Control ; 23(12): 2013-21, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23073789

ABSTRACT

PURPOSE: In the absence of gold standard diagnoses, we estimate age-specific false-positive and false-negative prediction rates of HPV-, cytology-, and histology-based tests for significant cervical lesions (SCL) in US women with AGC-NOS Pap smear diagnoses. METHODS: Modified Latent Class Model (LCM) analyses, with prevalence of SCL modeled as a function of age, were applied to GOG-0171 study data (n = 122). The accuracies of several HPV-based tests, including Hybrid Capture II high-risk HPV (HC2 H-HPV); carbonic anhydrase IX (CA-IX); and invasive histological diagnosis, were compared. 1-PPV and 1-NPV were written as functions of sensitivity, specificity, and prevalence to obtain age-specific false-positive and false-negative rates. RESULTS: The histology-based test was nearly perfect (sensitivity = 1.00, CI = 0.98-1.00; specificity = 0.99, CI = 0.96-1.00). Otherwise, HC2 H-HPV performed best (sensitivity = 1.00, CI = 1.00-1.00; specificity = 0.87, CI = 0.79-0.94). The false-positive detection rates (1-PPV) for HC2 H-HPV were high (>17 %) at each age, while those of the histological diagnoses were low (<5 % at ages ≤60 and <17 % overall ages). False-negative prediction rates (1-NPV) for HC2 H-HPV were <0.11 % at each age and were uniformly lower than those of other tests, including the histology-based test (<0.25 %). CA-IX together with HC2 H-HPV did not improve performance. CONCLUSIONS: Women with negative HC2 H-HPV can safely forego invasive treatment (i.e., cone or LEEP biopsy, hysterectomy) in favor of observational follow-up. Additional biomarkers must be found for use in combination with HC2 H-HPV to reduce false-positive rates. This novel application of a modified LCM exemplifies methods for potential use in future cancer screening studies when gold standard diagnoses are not available.


Subject(s)
Papillomaviridae/isolation & purification , Papillomavirus Infections/diagnosis , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/virology , Adult , Aged , Cytodiagnosis , Early Detection of Cancer , Female , Humans , Mass Screening/methods , Middle Aged , Papanicolaou Test , Papillomaviridae/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Sensitivity and Specificity , Vaginal Smears , Young Adult , Uterine Cervical Dysplasia/pathology
6.
J Ethnopharmacol ; 144(2): 453-6, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23022321

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Microenvironmental conditions contribute towards varying cellular responses to plant extract treatments. Hypoxic cancer cells are known to be resistant to radio- and chemo-therapy. New therapeutic strategies specifically targeting these cells are needed. Plant extracts used in Traditional Chinese Medicine (TCM) can offer promising candidates. Despite their widespread usage, information on their effects in hypoxic conditions is still lacking. In this study, we examined the cytotoxicity of a series of known TCM plant extracts under normoxic versus hypoxic conditions. MATERIALS AND METHODS: Pereskia grandifolia, Orthosiphon aristatus, Melastoma malabathricum, Carica papaya, Strobilanthes crispus, Gynura procumbens, Hydrocotyle sibthorpioides, Pereskia bleo and Clinacanthus nutans leaves were dried, blended into powder form, extracted in methanol and evaporated to produce crude extracts. Human Saos-2 osteosarcoma cells were treated with various concentrations of the plant extracts under normoxia or hypoxia (0.5% oxygen). 24h after treatment, an MTT assay was performed and the IC(50) values were calculated. Effect of the extracts on hypoxia inducible factor (HIF) activity was evaluated using a hypoxia-driven firefly luciferase reporter assay. RESULTS: The relative cytotoxicity of each plant extract on Saos-2 cells was different in hypoxic versus normoxic conditions. Hypoxia increased the IC(50) values for Pereskia grandifola and Orthosiphon aristatus extracts, but decreased the IC(50) values for Melastoma malabathricum and Carica papaya extracts. Extracts of Strobilanthes crispus, Gynura procumbens, Hydrocotyle sibthorpioides had equivalent cytotoxic effects under both conditions. Pereskia bleo and Clinacanthus nutans extracts were not toxic to cells within the concentration ranges tested. The most interesting result was noted for the Carica papaya extract, where its IC(50) in hypoxia was reduced by 3-fold when compared to the normoxic condition. This reduction was found to be associated with HIF inhibition. CONCLUSION: Hypoxia variably alters the cytotoxic effects of TCM plant extracts on cancer cells. Carica papaya showed enhanced cytotoxic effect on hypoxic cancer cells by inhibiting HIF activities. These findings provide a plausible approach to killing hypoxic cancer cells in solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1/metabolism , Medicine, Chinese Traditional , Plant Extracts/pharmacology , Cell Line, Tumor , Cell Survival , Humans , Plants, Medicinal
7.
Virol J ; 9: 155, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22877087

ABSTRACT

Enterovirus 71 (EV71) causes severe neurological diseases resulting in high mortality in young children worldwide. Development of an effective vaccine against EV71 infection is hampered by the lack of appropriate animal models for efficacy testing of candidate vaccines. Previously, we have successfully tested the immunogenicity and protectiveness of a candidate EV71 vaccine, containing recombinant Newcastle disease virus capsids that display an EV71 VP1 fragment (NPt-VP11-100) protein, in a mouse model of EV71 infection. A drawback of this system is its limited window of EV71 susceptibility period, 2 weeks after birth, leading to restricted options in the evaluation of optimal dosing regimens. To address this issue, we have assessed the NPt-VP11-100 candidate vaccine in a hamster system, which offers a 4-week susceptibility period to EV71 infection. Results obtained showed that the NPt-VP11-100 candidate vaccine stimulated excellent humoral immune response in the hamsters. Despite the high level of antibody production, they failed to neutralize EV71 viruses or protect vaccinated hamsters in viral challenge studies. Nevertheless, these findings have contributed towards a better understanding of the NPt-VP11-100 recombinant protein as a candidate vaccine in an alternative animal model system.


Subject(s)
Antibodies, Viral/blood , Capsid Proteins/immunology , Enterovirus A, Human/immunology , Immunization/methods , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Capsid Proteins/administration & dosage , Cricetinae , Disease Models, Animal , Enterovirus Infections/prevention & control , Mesocricetus , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage
8.
Cancer Lett ; 325(1): 89-98, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22743615

ABSTRACT

This study identified LTBP-2 as a pleiotropic tumor suppressor in nasopharyngeal carcinoma, which safeguards against critical malignant behaviors of tumor cells. LTBP-2 expression was significantly decreased or lost in up to 100% of NPC cell lines (7/7) and 80% of biopsies (24/30). Promoter hypermethylation was found to be involved in LTBP-2 silencing. Using a tetracycline-regulated inducible expression system, we unveiled functional roles of LTBP-2 in suppressing colony formation, anchorage-independent growth, cell migration, angiogenesis, VEGF secretion, and tumorigenicity. Three-dimensional culture studies suggested the involvement of LTBP-2 in maintenance of tumor cell dormancy in a growth factor favorable microenvironment.


Subject(s)
Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Nasopharyngeal Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Carcinoma , Cell Line, Tumor , Cell Movement/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Methylation , Decitabine , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Humans , Hydroxamic Acids/pharmacology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/blood supply , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Promoter Regions, Genetic , Tumor Microenvironment , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Int J Cancer ; 130(1): 83-95, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21387288

ABSTRACT

Suppressive effects of DUSP6 in tumorigenesis and EMT-associated properties were observed. Dual-specificity phosphatase (DUSP6) is a MAP kinase phosphatase (MKP) negatively regulating the activity of ERK, one of the major molecular switches in the MAPK signaling cascade propagating the signaling responses during malignancies. The impact of DUSP6 in EMT and its contribution to tumor dissemination has not yet been characterized. Due to differences in tumor microenvironments affecting cell signaling during cancer progression, DUSP6 may play varying roles in tumor development. We sought to examine the potential role of DUSP6-mediated tumorigenesis and EMT-associated properties in two aerodigestive tract cancers, namely, esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). Significant loss of DUSP6 was observed in 100% of 11 ESCC cell lines and 71% of seven NPC cell lines. DUSP6 expression was down-regulated in 40% of 30 ESCC tumor tissues and 75% of 20 NPC tumor tissues compared to their respective normal counterparts. Suppressive effects of DUSP6 in tumor formation and cancer cell mobility are seen in in vivo tumorigenicity assay and in vitro colony formation, three-dimensional Matrigel culture, cell migration and invasion chamber tests. Notably, overexpression of DUSP6 impairs EMT-associated properties. Furthermore, tissue microarray analysis reveals a clinical association of DUSP6 expression with better patient survival. Taken together, our study provides a novel insight into understanding the functional impact of DUSP6 in tumorigenesis and metastasis of ESCC and NPC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Movement , Dual Specificity Phosphatase 6/metabolism , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/pathology , Nasopharyngeal Neoplasms/pathology , Animals , Blotting, Western , Carcinoma , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA Methylation , Dual Specificity Phosphatase 6/genetics , Epigenomics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Staging , Phenotype , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Tissue Array Analysis
10.
Cancer Lett ; 316(1): 39-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154084

ABSTRACT

The group 2 LIM domain protein, Cysteine-rich intestinal protein 2 (CRIP2) was found to play an important role in esophageal squamous cell carcinoma (ESCC) tumorigenesis. Subcellular fractionation studies show that CRIP2 is expressed in the nucleus. Real-time quantitative PCR shows CRIP2 expression is down-regulated in ESCC tissues and cell lines. Functional studies reveal that CRIP2 reduces colony formation, growth, and invasion abilities. Furthermore, over-expression of CRIP2 induces apoptosis through induction of active caspases 3 and 9 proteins. In conclusion, this study shows CRIP2 plays an important role in the development of ESCC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Real-Time Polymerase Chain Reaction/methods , Up-Regulation
11.
Semin Cancer Biol ; 22(2): 87-95, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22154888

ABSTRACT

The identification of cancer genes in sporadic cancers has been recognized as a major challenge in the field. It is clear that deletion mapping, genomic sequencing, comparative genomic hybridization, or global gene expression profiling alone would not have easily identified candidate tumor suppressor genes (TSGs) from the huge array of lost regions or genes observed in nasopharyngeal carcinoma (NPC). In addition, the epigenetically silenced genes would not have been recognized by the mapping of deleted regions. In this review, we describe how functional approaches using monochromosome transfer may be used to circumvent the above problems and identify TSGs in NPC. A few examples of selected NPC TSGs and their functional roles are reviewed. They regulate a variety of gene functions including cell growth and proliferation, adhesion, migration, invasion, epithelial-mesenchymal transition, metastasis, and angiogenesis. These studies show the advantages of using functional approaches for identification of TSGs.


Subject(s)
Chromosome Mapping/methods , Genetic Predisposition to Disease/genetics , Nasopharyngeal Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Carcinoma , Cell Fusion/methods , Cell Movement/genetics , Humans , Hybrid Cells/metabolism , Mice , Nasopharyngeal Carcinoma
12.
J Med Virol ; 83(10): 1783-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21837796

ABSTRACT

Enterovirus 71 (EV71) infection may cause severe neurological complications, particularly in young children. Despite the risks, there are still no commercially available EV71 vaccines. Hence, a candidate vaccine construct, containing recombinant Newcastle disease virus capsids that display an EV71 VP1 fragment (NPt-VP1(1-100) ) protein, was evaluated in a mouse model of EV71 infection. Previously, it was shown that this protein construct provoked a strong immune response in vaccinated adult rabbits. That study, however, did not address the issue of its effectiveness against EV71 infection in young animals. In the present study, EV71 viral challenge in vaccinated newborn mice resulted in more than 40% increase in survival rate. Significantly, half of the surviving mice fully recovered from their paralysis. Histological analysis of all of the surviving mice revealed a complete clearance of EV71 viral antigens from their brains and spinal cords. In hind limb muscles, the amounts of the antigens detected correlated with the degrees of tissue damage and paralysis. Findings from this study provide evidence that immunization with the NPt-VP1(1-100) immunogen in a newborn mouse model confers partial protection against EV71 infection, and also highlights the importance of NPt-VP1(1-100) as a possible candidate vaccine for protection against EV71 infections.


Subject(s)
Capsid Proteins/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/prevention & control , Newcastle disease virus/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Antigens, Viral/immunology , Capsid/immunology , Chlorocebus aethiops , Cytokines/blood , Enterovirus A, Human/genetics , Enterovirus Infections/immunology , Immunization , Mice , Neutralization Tests , Newcastle disease virus/genetics , Vaccines, Synthetic/immunology , Vero Cells
13.
Proc Natl Acad Sci U S A ; 108(20): 8390-5, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21540330

ABSTRACT

Chromosome 14 was transferred into tumorigenic nasopharyngeal carcinoma and esophageal carcinoma cell lines by a microcell-mediated chromosome transfer approach. Functional complementation of defects present in the cancer cells suppressed tumor formation. A candidate tumor-suppressor gene, cysteine-rich intestinal protein 2 (CRIP2), located in the hot spot for chromosomal loss at 14q32.3, was identified as an important candidate gene capable of functionally suppressing tumor formation. Previous studies have shown that CRIP2 is associated with development. To date, no report has provided functional evidence supporting a role for CRIP2 in tumor development. The present study provides unequivocal evidence that CRIP2 can functionally suppress tumorigenesis. CRIP2 is significantly down-regulated in nasopharyngeal carcinoma cell lines and tumors. CRIP2 reexpression functionally suppresses in vivo tumorigenesis and angiogenesis; these effects are induced by its transcription-repressor capability. It interacts with the NF-κB/p65 to inhibit its DNA-binding ability to the promoter regions of the major proangiogenesis cytokines critical for tumor progression, including IL6, IL8, and VEGF. In conclusion, we provide compelling evidence that CRIP2 acts as a transcription repressor of the NF-κB-mediated proangiogenic cytokine expression and thus functionally inhibits tumor formation and angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Transformation, Neoplastic/genetics , Cytokines/genetics , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Angiogenic Proteins/analysis , Cell Line , Cell Line, Tumor , Chromosomes, Human, Pair 14 , Cytokines/physiology , Humans , LIM Domain Proteins , Repressor Proteins/physiology
14.
Int J Cancer ; 129(3): 565-73, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-20878956

ABSTRACT

Our previous studies of chromosome 14 transfer into tumorigenic esophageal squamous cell carcinoma (ESCC) cell line, SLMT, suggested the existence of tumor suppressor genes on chromosome 14. Gene expression profiling of microcell hybrids and the tumor segregants identified an interesting gene, LTBP-2 (latent transforming growth factor ß binding protein 2), which has been analyzed here for its role in ESCC. LTBP-2 maps to 14q24 and encodes a secreted protein, which is a component of the extracellular matrix microfibrils. LTBP-2 expression was downregulated in ESCC cell lines and tumor tissues. Promoter hypermethylation was found to be involved in LTBP-2 inactivation. Functional studies indicated its tumor-suppressive roles in ESCC. In the in vitro colony formation and Matrigel three-dimensional culture assays, LTBP-2 decreased the colony-forming abilities of ESCC cell lines. LTBP-2 expression was associated with reduction of cell migrating and invasive abilities. LTBP-2 could also reduce the tube-forming ability of endothelial cells. Moreover, LTBP-2 induced tumor suppression in in vivo nude mouse assays. Tissue microarray immunohistochemical staining analysis indicated that LTBP-2 expression is reduced in tumor tissues when compared to normal tissues, and LTBP-2 expression correlated significantly with the survival of ESCC patients. Thus, LTBP-2 appears to play an important role in ESCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Latent TGF-beta Binding Proteins/metabolism , Animals , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , DNA Methylation , Down-Regulation , Esophageal Neoplasms/mortality , Female , Genes, Tumor Suppressor , Humans , Latent TGF-beta Binding Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Tumor Stem Cell Assay
15.
Int J Cancer ; 129(8): 1826-37, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21165953

ABSTRACT

The association of Matrix metalloproteinase-19 (MMP19) in the development of nasopharyngeal carcinoma (NPC) was identified from differential gene profiling, which showed MMP19 was one of the candidate genes down-regulated in the NPC cell lines. In this study, quantitative RT-PCR and Western blot analysis showed MMP19 was down-regulated in all seven NPC cell lines. By tissue microarray immunohistochemical staining, MMP19 appears down-regulated in 69.7% of primary NPC specimens. Allelic deletion and promoter hypermethylation contribute to MMP19 down-regulation. We also clearly demonstrate that the catalytic activity of MMP19 plays an important role in antitumor and antiangiogenesis activities in comparative studies of the wild-type and the catalytically inactive mutant MMP19. In the in vivo tumorigenicity assay, only the wild-type (WT), but not mutant, MMP19 transfectants suppress tumor formation in nude mice. In the in vitro colony formation assay, WT MMP19 dramatically reduces colony-forming ability of NPC cell lines, when compared to the inactive mutant. In the tube formation assay of human umbilical vein endothelial cells and human microvascular endothelial cells (HMEC-1), secreted WT MMP19, but not mutant MMP19, induces reduction of tube-forming ability in endothelial cells with decreased vascular endothelial growth factor (VEGF) in conditioned media detected by enzyme-linked immunosorbent assay (ELISA). The anti-angiogenic activity of WT MMP19 is correlated with suppression of tumor formation. These results now clearly show that catalytic activity of MMP19 is essential for its tumor suppressive and anti-angiogenic functions in NPC.


Subject(s)
Matrix Metalloproteinases, Secreted/physiology , Nasopharyngeal Neoplasms/metabolism , Angiogenesis Inhibitors , Animals , Carcinoma , Catalysis , Cell Line, Tumor , DNA Methylation , Down-Regulation , Humans , Loss of Heterozygosity , Matrix Metalloproteinases, Secreted/genetics , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Transfection
16.
Cancer Res ; 70(14): 6004-14, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20570892

ABSTRACT

Activation of the mitogen-activated protein kinase (MAPK) pathway plays a major role in neoplastic cell transformation. Using a proteomics approach, we identified alpha tubulin and beta tubulin as proteins that interact with activated MAP/extracellular signal-regulated kinase kinase 1 (MEK1), a central MAPK regulatory kinase. Confocal analysis revealed spatiotemporal control of MEK1-tubulin colocalization that was most prominent in the mitotic spindle apparatus in variant HT1080 human fibrosarcoma cells. Peptide arrays identified the critical role of positively charged amino acids R108, R113, R160, and K157 on the surface of MEK1 for tubulin interaction. Overexpression of activated MEK1 caused defects in spindle arrangement, chromosome segregation, and ploidy. In contrast, chromosome polyploidy was reduced in the presence of an activated MEK1 mutant (R108A, R113A) that disrupted interactions with tubulin. Our findings indicate the importance of signaling by activated MEK1-tubulin in spindle organization and chromosomal instability.


Subject(s)
Fibrosarcoma/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Tubulin/metabolism , Actins/metabolism , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Fibrosarcoma/enzymology , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Humans , Immunoprecipitation , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Mutation , Ploidies , Spindle Apparatus/metabolism
17.
Cancer Res ; 70(13): 5567-76, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20551050

ABSTRACT

ADAMTS metalloprotease family member ADAMTS9 maps to 3p14.2 and shows significant associations with the aerodigestive tract cancers esophageal squamous cell carcinoma (ESCC) and nasopharyngeal carcinoma (NPC). However, the functional impact of ADAMTS9 on cancer development has not been explored. In this study, we evaluated the hypothesized antiangiogenic and tumor-suppressive functions of ADAMTS9 in ESCC and NPC, in stringent tumorigenicity and Matrigel plug angiogenesis assays. ADAMTS9 activation suppressed tumor formation in nude mice. Conversely, knockdown of ADAMTS9 resulted in clones reverting to the tumorigenic phenotype of parental cells. In vivo angiogenesis assays revealed a reduction in microvessel numbers in gel plugs injected with tumor-suppressive cell transfectants. Similarly, conditioned medium from cell transfectants dramatically reduced the tube-forming capacity of human umbilical vein endothelial cells. These activities were associated with a reduction in expression levels of the proangiogenic factors MMP9 and VEGFA, which were consistently reduced in ADAMTS9 transfectants derived from both cancers. Taken together, our results indicate that ADAMTS9 contributes an important function in the tumor microenvironment that acts to inhibit angiogenesis and tumor growth in both ESCC and NPC.


Subject(s)
ADAM Proteins/biosynthesis , Esophageal Neoplasms/blood supply , Esophageal Neoplasms/enzymology , Nasopharyngeal Neoplasms/blood supply , Nasopharyngeal Neoplasms/enzymology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAMTS9 Protein , Animals , Down-Regulation , Endothelial Cells/cytology , Enzyme Activation , Esophageal Neoplasms/genetics , Female , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/genetics , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Transfection , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
18.
J Biol Chem ; 285(20): 15511-15522, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20215110

ABSTRACT

CADM1 encodes a multifunctional immunoglobulin-like cell adhesion molecule whose cytoplasmic domain contains a type II PSD95/Dlg/ZO-1 (PDZ)-binding motif (BM) for associating with other intracellular proteins. Although CADM1 lacks expression in T lymphocytes of healthy individuals, it is overexpressed in adult T-cell leukemia-lymphoma (ATL) cells. It has been suggested that the expression of CADM1 protein promotes infiltration of leukemic cells into various organs and tissues, which is one of the frequent clinical manifestations of ATL. Amino acid sequence alignment revealed that Tiam1 (T-lymphoma invasion and metastasis 1), a Rac-specific guanine nucleotide exchange factor, has a type II PDZ domain similar to those of membrane-associated guanylate kinase homologs (MAGUKs) that are known to bind to the PDZ-BM of CADM1. In this study, we demonstrated that the cytoplasmic domain of CADM1 directly interacted with the PDZ domain of Tiam1 and induced formation of lamellipodia through Rac activation in HTLV-I-transformed cell lines as well as ATL cell lines. Our results indicate that Tiam1 integrates signals from CADM1 to regulate the actin cytoskeleton through Rac activation, which may lead to tissue infiltration of leukemic cells in ATL patients.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Human T-lymphotropic virus 1/pathogenicity , Immunoglobulins/metabolism , Leukemia, T-Cell/pathology , Membrane Proteins/metabolism , Neoplasm Invasiveness , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Base Sequence , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Cell Line, Tumor , Guanine Nucleotide Exchange Factors/chemistry , Humans , Immunoglobulins/chemistry , Immunohistochemistry , Membrane Proteins/chemistry , Microscopy, Confocal , Molecular Sequence Data , Protein Binding , RNA Interference , RNA, Small Interfering , Sequence Homology, Amino Acid , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Tumor Suppressor Proteins/chemistry
19.
Gynecol Oncol ; 116(3): 452-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19913895

ABSTRACT

OBJECTIVES: This study aimed to determine whether carbonic anhydrase-IX (CA-IX) was associated with progression-free survival (PFS) and overall survival (OS) in women with high-risk, early-stage cervical cancer treated with adjuvant pelvic radiotherapy with or without radiosensitizing chemotherapy. METHODS: CA-IX expression was detected using an immunohistochemistry assay and categorized as low when 80% tumor cells display CA-IX staining. Associations between CA-IX expression and clinical characteristics, angiogenesis marker expression, and clinical outcome were evaluated. RESULTS: High CA-IX expression was observed in 35/166 (21.1%) of cases. CA-IX expression was not associated with age, race, stage, cell type, grade, positive margins, parametrial extensions, positive lymph nodes, or lymphovascular space invasion but was associated with tumor size categorized as <2 , 2-2.9 , or >or=3 cm (high expression: 4.7% vs. 23.2% vs. 32.5%, P=0.003) and cervical invasion confined to the inner two-thirds compared with the outer third of the cervix (high expression: 6.1% vs. 23.7%, P=0.028). CA-IX expression was not associated with immunohistochemical expression of p53, CD31, CD105, thrombospondin-1, or vascular endothelial growth factor-A. Women with high versus low CA-IX expression had similar PFS (P=0.053) and significantly worse OS (P=0.044). After adjusting for prognostic clinical covariates, high CA-IX expression was an independent prognostic factor for PFS (hazard ratio [HR]=2.12; 95% confidence interval [CI]=1.13-3.95; P=0.019) and OS (HR=2.41; 95% CI=1.24-4.68; P=0.009). CONCLUSIONS: Tumor hypoxia measured by immunohistochemical expression of CA-IX is an independent prognostic factor for both PFS and OS in high-risk, early-stage cervical cancer.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carbonic Anhydrases/biosynthesis , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/therapy , Adult , Aged , Carbonic Anhydrase IX , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Proportional Hazards Models , Risk Factors , Survival Rate , Uterine Cervical Neoplasms/pathology
20.
Int J Cancer ; 127(2): 304-12, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-19921696

ABSTRACT

THY1 was previously identified as a candidate tumor suppressor gene (TSG) associated with lymph node metastases in nasopharyngeal carcinoma (NPC) through functional studies. It was identified by oligonucleotide microarray analysis as an interesting differentially expressed gene. However, direct functional evidence is still lacking for THY1 being a TSG in NPC, as in vivo tumorigenicity assays have not been previously reported in our last study of THY1. In this study, a tetracycline-inducible expression vector, pETE-Bsd, was used to obtain stable transfectants of THY1. The stringent in vivo tumorigenicity assay results show that the activation of THY1 suppresses tumor formation of HONE1 cells in nude mice, and the tumor formation ability was restored in the presence of doxycycline (a tetracycline analog), when the gene is shut off. Functional inactivation of this gene is observed in all the tumors derived from the tumorigenic transfectant. The tumor suppressive effect could be repressed by knockdown of THY1 expression in nontumorigenic microcell hybrids. Further studies indicate that expression of THY1 inhibits HONE1 cell growth in vitro by arresting cells in G(0)/G(1) phase. It greatly reduces the ability for anchorage-independent growth. The invasiveness of HONE1 cells was also inhibited by the expression of THY1. These findings suggest that THY1 is a TSG in NPC, which is involved in invasion and shows an association with tumor metastasis. Taken together, THY1 clearly plays an important functional role in tumor suppression in NPC.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor/physiology , Nasopharyngeal Neoplasms/pathology , Thy-1 Antigens/physiology , Animals , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Proliferation , Female , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...