Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomark Res ; 11(1): 70, 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37468977

ABSTRACT

Biomarkers are detectable molecules that can reflect specific physiological states of cells, organs, and organisms and therefore be regarded as indicators for specific diseases. And the discovery of biomarkers plays an essential role in cancer management from the initial diagnosis to the final treatment regime. Practically, reliable clinical biomarkers are still limited, restricted by the suboptimal methods in biomarker discovery. Nucleic acid aptamers nowadays could be used as a powerful tool in the discovery of protein biomarkers. Nucleic acid aptamers are single-strand oligonucleotides that can specifically bind to various targets with high affinity. As artificial ssDNA or RNA, aptamers possess unique advantages compared to conventional antibodies. They can be flexible in design, low immunogenicity, relative chemical/thermos stability, as well as modifying convenience. Several SELEX (Systematic Evolution of Ligands by Exponential Enrichment) based methods have been generated recently to construct aptamers for discovering new biomarkers in different cell locations. Secretome SELEX-based aptamers selection can facilitate the identification of secreted protein biomarkers. The aptamers developed by cell-SELEX can be used to unveil those biomarkers presented on the cell surface. The aptamers from tissue-SELEX could target intracellular biomarkers. And as a multiplexed protein biomarker detection technology, aptamer-based SOMAScan can analyze thousands of proteins in a single run. In this review, we will introduce the principle and workflow of variations of SELEX-based methods, including secretome SELEX, ADAPT, Cell-SELEX and tissue SELEX. Another powerful proteome analyzing tool, SOMAScan, will also be covered. In the second half of this review, how these methods accelerate biomarker discovery in various diseases, including cardiovascular diseases, cancer and neurodegenerative diseases, will be discussed.

2.
Front Cell Dev Biol ; 10: 1053984, 2022.
Article in English | MEDLINE | ID: mdl-36544906

ABSTRACT

Aptamers are short single-strand oligonucleotides that can form secondary and tertiary structures, fitting targets with high affinity and specificity. They are so-called "chemical antibodies" and can target specific biomarkers in both diagnostic and therapeutic applications. Systematic evolution of ligands by exponential enrichment (SELEX) is usually used for the enrichment and selection of aptamers, and the targets could be metal ions, small molecules, nucleotides, proteins, cells, or even tissues or organs. Due to the high specificity and distinctive binding affinity of aptamers, aptamer-drug conjugates (ApDCs) have demonstrated their potential role in drug delivery for cancer-targeting therapies. Compared with antibodies which are produced by a cell-based bioreactor, aptamers are chemically synthesized molecules that can be easily conjugated to drugs and modified; however, the conventional ApDCs conjugate the aptamer with an active drug using a linker which may add more concerns to the stability of the ApDC, the drug-releasing efficiency, and the drug-loading capacity. The function of aptamer in conventional ApDC is just as a targeting moiety which could not fully perform the advantages of aptamers. To address these drawbacks, scientists have started using active nucleotide analogs as the cargoes of ApDCs, such as clofarabine, ara-guanosine, gemcitabine, and floxuridine, to replace all or part of the natural nucleotides in aptamer sequences. In turn, these new types of ApDCs, aptamer nucleotide analog drug conjugates, show the strength for targeting efficacy but avoid the complex drug linker designation and improve the synthetic efficiency. More importantly, these classic nucleotide analog drugs have been used for many years, and aptamer nucleotide analog drug conjugates would not increase any unknown druggability risk but improve the target tumor accumulation. In this review, we mainly summarized aptamer-conjugated nucleotide analog drugs in cancer-targeting therapies.

3.
Mol Ther Nucleic Acids ; 22: 153-165, 2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32927364

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-associated cancer characterized by a high degree of recurrence, angiogenesis, and metastasis. The importance of alternative pro-angiogenesis pathways including viral factors has emerged after decades of directly targeting various signaling components. Using NPC as a model, we identified an essential oncogenic pathway underlying angiogenesis regulation that involves the inhibition of a tumor suppressor, Spry3, and its downstream targets by EBV-miR-BART10-5p (BART10-5p) and hsa-miR-18a (miR-18a). Overexpression of EBV-miR-BART10-5p and hsa-miR-18a strongly promotes angiogenesis in vitro and in vivo by regulating the expression of VEGF and HIF1-α in a Spry3-dependent manner. In vitro or in vivo treatment with iRGD-tagged exosomes containing antagomiR-BART10-5p and antagomiR-18a preferentially suppressed the angiogenesis and growth of NPC. Our findings first highlight the role of EBV-miR-BART10-5p and oncogenic hsa-miR-18a in NPC angiogenesis and also shed new insights into the clinical intervention and therapeutic strategies for nasopharyngeal carcinoma and other virus-associated tumors.

4.
J Gen Virol ; 100(6): 999-1012, 2019 06.
Article in English | MEDLINE | ID: mdl-30816843

ABSTRACT

Epstein-Barr virus (EBV) infection is strongly associated with nasopharyngeal carcinoma, a common cancer in Southeast Asia and certain regions of Africa. However, the dynamics of EBV episome maintenance in infected nasopharyngeal epithelial (NPE) cells remain largely undefined. Here, we report the establishment of a highly efficient cell-free EBV infection method for NPE cells. By using this method, we have defined some of the dynamic events involved in the early stage of EBV infection in NPE cells. We report, for the first time, a rapid loss of EBV copies from infected NPE cells during the first 12-72 h post-infection. The rate of EBV loss slowed at later stages of infection. Live cell imaging revealed that the freshly infected NPE cells were delayed in entry into mitosis compared with uninfected cells. Freshly infected NPE cells transcribed significantly higher levels of lytic EBV genes BZLF1 and BMRF1 yet significantly lower levels of EBER1/2 than stably infected NPE cells. Notably, there were very low or undetectable levels of protein expressions of EBNA1, LMP1, Zta and Rta in freshly infected NPE cells, whereas EBNA1 and LMP1 proteins were readily detected in stable EBV-infected NPE cells. The kinetics of EBV loss and the differential EBV gene expression profiles between freshly and stably infected NPE cells are in line with the suggestion of epigenetic changes in the EBV genome that affect viral gene expression and the adaptation of host cells to EBV infection to maintain persistent EBV infection in NPE cells.


Subject(s)
Epithelial Cells/virology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Nasopharynx/virology , Cell Line , Epigenesis, Genetic/genetics , Epstein-Barr Virus Nuclear Antigens/genetics , Humans , Trans-Activators/genetics , Transcriptome/genetics
5.
Cancer Lett ; 343(1): 24-32, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24041865

ABSTRACT

The dual PI3K-mTOR inhibitor BEZ235 was evaluated in preclinical models of nasopharyngeal carcinoma (NPC). The IC50 value of BEZ235 for growth was in the nanomolar range in vitro, induce G1 cycle arrest and apoptosis, and inhibited AKT and mTOR signaling in most NPC cell lines. No synergistic effect was observed when BEZ235 was combined with chemotherapy. BEZ235 increased MAPK activation in vitro but not in vivo. A daily schedule was more effective than a weekly schedule on tumor growth and inhibition of downstream mTOR signaling in vivo. The activity of BEZ235 maybe independent of the PIK3CA amplification and mutation status.


Subject(s)
Gene Expression Regulation, Neoplastic , Imidazoles/pharmacology , Nasopharyngeal Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Quinolines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma , Cell Cycle , Cell Line, Tumor , Cell Survival , Cisplatin/pharmacology , Enzyme Activation , Female , Humans , Inhibitory Concentration 50 , MAP Kinase Signaling System , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Neoplasm Transplantation , Paclitaxel/pharmacology , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors
6.
Invest New Drugs ; 30(1): 48-58, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20694504

ABSTRACT

Recent studies revealed an unexpected role of the neurotrophin receptor pathway, BDNF/TrkB signaling, in cancer metastasis and anoikis (i.e. detachment-induced cell death). Survival of cancer cells in detached state (known as anoikis-resistance) is known to be pre-requisite for metastasis. Nasopharyngeal carcinoma (NPC), an endemic head and neck cancer in Southeast Asia, is highly invasive, metastatic, and etiologically associated with Epstein-Barr virus (EBV, an oncovirus) infection. Mechanistic studies on the invasive/metastatic nature of NPC can facilitate the development of anti-metastatic therapy in NPC. Thus far, the role of BDNF/TrkB signaling in virus-associated human cancer is unclear. Here, using multiple cell line models of NPC with EBV-association (HONE-1-EBV, HK1-LMP1 and C666-1), we investigated the potential involvement of BDNF/TrkB signaling in cellular migration and anoikis-resistant characteristics of NPC. We found that all three EBV-associated NPC cell lines tested were intrinsically anoikis-resistant (i.e. survived in detached state) and expressed both BDNF and TrkB. BDNF stimulation induced cellular migration, but not proliferation of these cells. Further, we examined if pharmacologic targeting of anoikis-resistance of NPC cells can be achievable by a proof-of-concept Trk inhibitor, K252a, in these EBV-associated NPC models. Our results demonstrated that K252a, was able to attenuate BDNF-induced migration and proliferation of NPC cells. More importantly, we demonstrated for the first time that K252a harbored potent anoikis-sensitization activity (i.e. sensitizing cancer cells to detachment-induced cell death) against EBV-associated human cancer cells, namely NPC cells. This proof-of-concept study demonstrated that K252a, a Trk inhibitor, can potentially be used as an anoikis-sensitizing agent in NPC.


Subject(s)
Anoikis/drug effects , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Cell Movement/drug effects , Herpesvirus 4, Human/pathogenicity , Indole Alkaloids/pharmacology , Nasopharyngeal Neoplasms/enzymology , Nasopharyngeal Neoplasms/virology , Protein Kinase Inhibitors/pharmacology , Receptor, trkB/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/metabolism , Carcinoma , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Receptor, trkB/metabolism , Signal Transduction/drug effects
7.
Invest New Drugs ; 29(3): 401-10, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20013349

ABSTRACT

Hypoxia is commonly developed in solid tumors, which contributes to metastasis as well as radio- and chemo-resistance. Nasopharyngeal carcinoma (NPC) is a highly invasive and metastatic head and neck cancer prevalent in Southeast Asia with a high incidence rate of 15-30/100,000 persons/year (comparable to that of pancreatic cancer in the US). Previous clinical studies in NPC showed that hypoxia is detected in almost 100% of primary tumors and overexpression of hypoxia markers correlated with poor clinical outcome. Tirapazamine (TPZ) is a synthetic hypoxia-activated prodrug, which preferentially forms cytotoxic and DNA-damaging free radicals under hypoxia, thus selectively eradicate hypoxic cells. Here, we hypothesized that specific hypoxia-targeting by this clinical trial agent may be therapeutic for NPC. Our findings demonstrated that under hypoxia, TPZ was able to induce preferential growth inhibition of NPC cells, which was associated with marked cell cycle arrest at S-phase and PARP cleavage (a hallmark of apoptosis). Examination of S-phase checkpoint regulators revealed that Chk1 and Chk2 were selectively activated by TPZ in NPC cells under hypoxia. Hypoxia-selectivity of TPZ was also demonstrated by preferential downregulation of several important hypoxia-induced markers (HIF-1α, CA IX and VEGF) under hypoxia. Furthermore, we demonstrated that TPZ was equally effective and hypoxia-selective even in the presence of the EBV oncoprotein, LMP1 or the EBV genome. In summary, encouraging results from this proof-of-concept study implicate the therapeutic potential of hypoxia-targeting approaches for the treatment of NPC.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Triazines/pharmacology , Apoptosis/drug effects , Carcinoma , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genome, Viral/genetics , Herpesvirus 4, Human/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/enzymology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Poly(ADP-ribose) Polymerases/metabolism , S Phase/drug effects , Signal Transduction/drug effects , Tirapazamine , Up-Regulation/drug effects , Viral Matrix Proteins/metabolism
8.
J Cancer Res Clin Oncol ; 137(3): 415-22, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20440510

ABSTRACT

AIMS: This study aimed at evaluating the potential anti-proliferative effects of the microRNA let-7 family in nasopharyngeal carcinoma (NPC) cells. In addition, the association between let-7 suppression and DNA hypermethylation is examined. MATERIALS AND METHODS: Levels of mature let-7 family members (-a, -b, -d, -e, -g, and -i) in normal nasopharyngeal cells (NP69 and NP460) and nasopharyngeal carcinoma cells (HK1 and HONE1) were measured by real-time quantitative PCR. Cell-proliferation assay and c-Myc immunohistochemical staining were performed on NPC cells transfected with let-7 precursor molecules. In addition, expression changes in let-7 family members in response to demethylating agents (5-azacytidine and zebularine) were also examined. RESULTS: In comparison with the normal nasopharyngeal cells, let-7 (-a, -b, -d, -e, -g, and -i) levels were reduced in nasopharyngeal carcinoma cells. Ectopic expression of the let-7 family in nasopharyngeal carcinoma cells resulted in inhibition of cell proliferation through downregulation of c-Myc expression. Demethylation treatment of nasopharyngeal carcinoma cells caused activation of let-7 expression in poorly differentiated nasopharyngeal carcinoma cells only. CONCLUSION: Our results suggested that miRNA let-7 might play a role in the proliferation of NPC. DNA methylation is a potential regulatory pathway, which is affected when let-7 is suppressed in NPC cells. However, the extent of DNA hypermethylation/hypomethylation in regulating let-7 expression requires further elucidation.


Subject(s)
MicroRNAs/genetics , Proto-Oncogene Proteins c-myc/genetics , Azacitidine/pharmacology , Carcinoma , Cell Growth Processes/genetics , Cell Line, Tumor , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Methylation/drug effects , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Proto-Oncogene Proteins c-myc/biosynthesis , Transfection , Up-Regulation/drug effects
9.
Integr Cancer Ther ; 9(4): 354-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21106616

ABSTRACT

HYPOTHESIS: Hepatocellular carcinoma (HCC) is one of the most malignant human tumors and one of the risk factors is its highly metastatic property. Coptidis Rhizoma aqueous extract (CRAE) is able to suppress the migration and invasion of HCC cells, MHCC97-L, and F-actin reorganization and Rho signaling inhibition is involved. MAIN METHODS: CRAE was prepared and analyzed by high-performance liquid chromatography combined with mass spectrometry. The cytotoxicity and antimigration action of CRAE on MHCC97-L cells were evaluated; Immunofluorescence and immunoblotting were used to investigate the proposed mechanism of CRAE action. KEY FINDINGS: Chemical analysis reveals that the active components in CRAE are berberine and berberine-like alkaloids. CRAE exhibits a significant inhibitory effect on MHCC97-L cell migration as indicated by wound-healing and invasion-chamber assays. No significant alteration of matrix metalloproteinases and urokinase-type plasminogen activator (uPA) expression were observed in MHCC97-L cells exposed to CRAE. Reduction of F-actin polymerization and damage to cytoskeleton network in MHCC97-L cells were observed after CRAE treatment. Furthermore, it was found that CRAE significantly downregulated the Rho/ROCK signaling pathway. SIGNIFICANCE: These results indicate that CRAE may act as a Rho/ROCK signaling inhibitor to suppress MHCC97-L cell migration in vitro and suggested that total alkaloids in Coptidis Rhizoma may be a potential agent for suppressing liver cancer invasion.


Subject(s)
Actins/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Drugs, Chinese Herbal/pharmacology , Liver Neoplasms/pathology , Protein Multimerization/drug effects , rhoA GTP-Binding Protein/antagonists & inhibitors , Actins/physiology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coptis chinensis , Drug Evaluation, Preclinical , Humans , Liver Neoplasms/metabolism , Neoplasm Invasiveness , Protein Multimerization/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , rhoA GTP-Binding Protein/metabolism
10.
J Cell Biochem ; 111(6): 1426-36, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20830746

ABSTRACT

Extensive studies have revealed that berberine, a small molecule derived from Coptidis rhizoma (Huanglian in Chinese) and many other plants, has strong anti-tumor properties. To better understand berberine-induced cell death and its underlying mechanisms in cancer, we examined autophagy and apoptosis in the human hepatic carcinoma cell lines HepG2 and MHCC97-L. The results of this study indicate that berberine can induce both autophagy and apoptosis in hepatocellular carcinoma cells. Berberine-induced cell death in human hepatic carcinoma cells was diminished in the presence of the cell death inhibitor 3-methyladenine, or following interference with the essential autophagy gene Atg5. Mechanistic studies showed that berberine may activate mitochondrial apoptosis in HepG2 and MHCC97-L cells by increasing Bax expression, the formation of permeable transition pores, cytochrome C release to cytosol, and subsequent activation of the caspases 3 and 9 execution pathway. Berberine may also induce autophagic cell death in HepG2 and MHCC97-L cells through activation of Beclin-1 and inhibition of the mTOR-signaling pathway by suppressing the activity of Akt and up-regulating P38 MAPK signaling. This is the first study to describe the role of Beclin-1 activation and mTOR inhibition in berberine-induced autophagic cell death. These results further demonstrate the potential of berberine as a therapeutic agent in the emerging list of cancer therapies with novel mechanisms.


Subject(s)
Berberine/pharmacology , Cell Survival/drug effects , Liver Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy/genetics , Beclin-1 , Berberine/therapeutic use , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cytochromes c/metabolism , Hep G2 Cells , Humans , Immunoblotting , Liver Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Med Virol ; 82(10): 1711-23, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20827769

ABSTRACT

Cell immortalization is regarded as an early and pre-requisite step in tumor development. Defining the specific genetic events involved in cell immortalization may provide insights into the early events of carcinogenesis. Nasopharyngeal carcinoma is common among the Southern Chinese population. Epstein-Barr virus (EBV) infection is associated closely with nasopharyngeal carcinoma. The involvement of LMP1 (an EBV-encoded oncogene) has been implicated in the pathogenesis of nasopharyngeal carcinoma. In this study, LMP1 expression, in combination with ectopic expression of hTERT (catalytic unit of human telomerase), was shown to extend the life span of primary cultures of nasopharyngeal epithelial cells and facilitate the immortalization of one of the cell lines (NP446). This is the first report on the successful immortalization of nasopharyngeal epithelial cells involving LMP1. The events associated with the immortalization of nasopharyngeal epithelial cells by LMP1/hTERT were characterized. Expression of c-Myc, Bmi-1, and Id-1 were upregulated at an early stage of immortalization. At a later stage of immortalization, downregulation of p21 and p16 expression were observed. Upregulation of EGFR expression and activation of MAPK signaling pathway were observed in LMP1/hTERT-immortalized nasopharyngeal epithelial cells. The LMP1/hTERT-immortalized NP446 cells were non-tumorigenic in immunosuppressed nude mice and retained anchorage-dependent growth, suggesting that additional events are required for tumorigenic transformation. The ability of the EBV-encoded LMP1, in the presence of hTERT expression, to extend the life span and immortalize primary cultures of nasopharyngeal epithelial cells supports the involvement of EBV infection and its viral products in the early stage of pathogenesis of nasopharyngeal carcinoma.


Subject(s)
Cell Transformation, Neoplastic , Epithelial Cells/cytology , Nasopharynx/cytology , Telomerase/biosynthesis , Viral Matrix Proteins/biosynthesis , Adult , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Male
12.
Carcinogenesis ; 30(12): 2085-94, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19843642

ABSTRACT

Nasopharyngeal carcinoma (NPC) is an Asian-prevalent head and neck cancer with high invasiveness. Although several important risk factors for NPC development have been identified, there is currently no preventive strategy for NPC, even in endemic regions. Signal transducer and activator of transcription 3 (STAT3) has been implicated in NPC carcinogenesis, which may serve as a potential target for cancer prevention. Here, we examined the chemopreventive potential of Cucurbitacin I, a natural-occurring selective inhibitor of JAK/STAT3, in NPC models. We hypothesized that Cucurbitacin I would prevent NPC invasion and tumor formation. Our data demonstrated that brief exposure of NPC cells to Cucurbitacin I was sufficient to significantly reduce the in vitro clonogenicity and in vivo tumorigenicity of NPC cells. The chemopreventive potential of Cucurbitacin I was further demonstrated by pre-dosing of the animals with Cucurbitacin I prior to tumor inoculation, which was found to be able to suppress tumor growth up to 7 days post-inoculation. The anti-proliferation activity of Cucurbitacin I was accompanied by downregulation of phospho-STAT3 and STAT3 target gene expression (e.g. cyclin D1 and Mcl-1). Cucurbitacin I also reduced the invasiveness of invasive NPC cell lines with elevated STAT3 activation. Furthermore, our data demonstrated for the first time that Cucurbitacin I harbored potent anoikis-sensitization activity (i.e. sensitizing cancer cells to detachment-induced cell death) against human cancer. Taken together, our results suggested that Cucurbitacin I may be a potent chemopreventive agent for NPC with anti-invasion and anoikis-sensitizing activities.


Subject(s)
Anoikis , Carcinoma/metabolism , Carcinoma/pathology , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Triterpenes/metabolism , Animals , Carcinoma/drug therapy , Cell Line, Tumor , Cell Survival , Collagen/chemistry , Drug Combinations , Female , Humans , Laminin/chemistry , Mice , Mice, Nude , Nasopharyngeal Neoplasms/drug therapy , Neoplasm Invasiveness , Neoplasm Transplantation , Proteoglycans/chemistry , STAT3 Transcription Factor/metabolism , Triterpenes/pharmacology
13.
Int J Cancer ; 125(8): 1884-93, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19588483

ABSTRACT

Nasopharyngeal cancer (NPC) is an Epstein-Barr virus (EBV)-associated head and neck cancer prevalent in Asia. Although with reasons not fully understood, the intrinsic invasiveness of NPC is believed to be EBV-linked. Recently, EBV was found to induce STAT3 activation. Constitutive STAT3 activation correlated with advanced clinical staging in NPC. We hypothesized that STAT3 activation by EBV directly contributes to the intrinsic invasiveness of NPC cells. Phospho-STAT3-Tyr705 was detected in high percentage of NPC tumors (7/10 cases). Using a paired NPC cell line model, HONE-1 and the EBV-infected counterpart, HONE-1-EBV, we found that HONE-1-EBV expressed a higher level of phospho-STAT3-Tyr705 and was approximately 11-fold more invasive than HONE-1. In HONE-1-EBV, STAT3 siRNA targeting inhibited both spontaneous and serum-induced invasion, as well as cell growth. Conversely, activation of STAT3 (by expressing an activated STAT3 mutant, namely STAT3C) in the parental HONE-1, mimicking EBV-induced STAT3 activation, significantly enhanced its invasiveness and proliferation, which was accompanied by increased expression of markers of mesenchymal status, proliferation and anti-apoptosis. Our results demonstrated that EBV-induced STAT3 activation is responsible for NPC cell proliferation and invasion. This was further confirmed by a small molecule inhibitor of JAK/STAT3, JSI-124. JSI-124 inhibited STAT3 activation in HONE-1-EBV, with subsequent growth inhibition, induction of PARP cleavage, abrogation of anchorage-independent growth and invasion. We found that EBV-independent activation of STAT3 by a growth factor, EGF, also contributed to NPC invasion. In conclusion, EBV-induced STAT3 activation directly contributes to the intrinsic invasiveness of NPC cells and STAT3 targeting may be beneficial in treating aggressive NPC.


Subject(s)
Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , Nasopharyngeal Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Collagen/metabolism , Colony-Forming Units Assay , Drug Combinations , Epidermal Growth Factor/pharmacology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Humans , Laminin/metabolism , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/virology , Neoplasm Invasiveness , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Tumor Cells, Cultured
14.
J Leukoc Biol ; 78(1): 239-47, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15800027

ABSTRACT

Intercellular adhesion molecule-1 (ICAM-1) has been shown to play crucial roles in mast cell interaction with other inflammatory cells and recruitment into the inflamed tissue. In the present study, human mast cell line-1 (HMC-1) was stimulated with different cytokines including stem cell factor (SCF), tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-13, IL-18, and IL-25. Cell-surface expression of ICAM-1 was assessed by flow cytometry. To elucidate the intracellular signal transduction regulating the ICAM-1 expression, phosphorylated extracellular signal-regulated kinase (ERK), phosphorylated p38 mitogen-activated protein kinase (MAPK), and nuclear factor (NF)-kappaB translocation were assessed by enzyme-linked immunosorbent assay. Results showed that SCF, TNF-alpha, and IL-13 but not IL-18 and IL-25 could up-regulate the surface expression of ICAM-1 on HMC-1 cells. A synergistic effect of SCF and TNF-alpha on ICAM-1 expression was demonstrated. This synergistic effect was shown to be dose-dependently enhanced by SCF but not TNF-alpha. Results indicated that SCF activated ERK, and TNF-alpha activated the p38 MAPK and NF-kappaB pathway. Selective inhibitor of ERK, PD098059, and c-kit inhibitors, STI571 and PP1, suppressed the combined SCF and TNF-alpha-induced ICAM-1 expression. BAY117082 but not SB203580, which are the inhibitors of NF-kappaB and p38 MAPK, respectively, suppressed the TNF-alpha-induced ICAM-1 expression. Therefore, SCF and TNF-alpha acted through ERK and the NF-kappaB pathway to regulate the ICAM-1 expression and elicited the synergistic effect. In conclusion, our results provide insight for cross-talk between different signaling pathways that can help in understanding the fine control of adhesion molecule expression under the concerted effects of cytokines.


Subject(s)
Cell Membrane/immunology , Intercellular Adhesion Molecule-1/immunology , Mast Cells/immunology , Stem Cell Factor/immunology , Tumor Necrosis Factor-alpha/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Communication/drug effects , Cell Communication/immunology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Inflammation/immunology , Inflammation/physiopathology , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/metabolism , Interleukin-13/immunology , Interleukin-13/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mast Cells/drug effects , Mast Cells/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Stem Cell Factor/pharmacology , Tumor Necrosis Factor-alpha/drug effects , Up-Regulation/drug effects , Up-Regulation/immunology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...