Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 493(4): 1498-1503, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28988106

ABSTRACT

Pancreatic adenocarcinoma upregulated factor (PAUF) overexpressed in pancreatic ductal adenocarcinoma (PDAC) plays a major role in tumor progression and metastasis by autocrine and paracrine manners. However, underlying molecular mechanism of PAUF functioning in pancreatic cancer are not fully understood yet. The objective of this study was to evaluate the potential of demilune cell and parotid protein 1 (DCPP1) as a putative mouse ortholog of human PAUF by sequence alignment and functional studies. Overexpression of mouse DCPP1 in Chinese hamster ovary (CHO) cells or pancreatic cancer cells increased cell proliferation, migration, invasion, and adhesion ability in vitro. Treatment of human pancreatic cancer cells with recombinant mouse DCPP1 elevated cell growth, motility, invasiveness, and adhesiveness. Mouse DCPP1 exerted its function on pancreatic cancer cells by activating intracellular signaling pathways involved in aggressive cancer phenotype of human pancreatic cancer cells. Moreover, subcutaneous injection of mice with DCPP1-overexpressing CHO cells increased tumor sizes. Taken together, we conclude that mouse DCPP1 is a multifunctional promoter of tumor growth through functional activation of pancreatic cancer cells, suggesting it to be an ortholog of human PAUF.


Subject(s)
Carcinoma, Pancreatic Ductal/physiopathology , Lectins/physiology , Pancreatic Neoplasms/physiopathology , Pregnancy Proteins/physiology , Animals , CHO Cells , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cricetulus , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins , Lectins/antagonists & inhibitors , Lectins/genetics , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pregnancy Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Microenvironment , Up-Regulation
2.
Oncotarget ; 7(31): 50315-50332, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27385093

ABSTRACT

TMPRSS4 is a novel type II transmembrane serine protease found at the cell surface that is highly expressed in pancreatic, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates tumor cell invasion, migration, and metastasis. We also found that TMPRSS4 activates the transcription factor activating protein-1 (AP-1) to induce cancer cell invasion. Here, we explored TMPRSS4-mediated cellular functions and the underlying mechanisms. TMPRSS4 induced Slug, an epithelial-mesenchymal transition (EMT)-inducing transcription factor, and cyclin D1 through activation of AP-1, composed of c-Jun and activating transcription factor (ATF)-2, which resulted in enhanced invasion and proliferation of PC3 prostate cancer cells. In PC3 cells, not only c-Jun but also Slug was required for TMPRSS4-mediated proliferation and invasion. Interestingly, Slug induced phosphorylation of c-Jun and ATF-2 to activate AP-1 through upregulation of Axl, establishing a positive feedback loop between Slug and AP-1, and thus induced cyclin D1, leading to enhanced proliferation. Using data from The Cancer Genome Atlas, we found that Slug expression positively correlated with that of c-Jun and cyclin D1 in human prostate cancers. Expression of Slug was positively correlated with that of cyclin D1 in various cancer cell lines, whereas expression of other EMT-inducing transcription factors was not. This study demonstrates that TMPRSS4 modulates both invasion and proliferation via Slug and cyclin D1, which is a previously unrecognized pathway that may regulate metastasis and cancer progression.


Subject(s)
Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Prostatic Neoplasms/metabolism , Serine Endopeptidases/metabolism , Snail Family Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Small Interfering/metabolism , Skin Neoplasms/metabolism , Up-Regulation
3.
J Biotechnol ; 205: 47-58, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25617682

ABSTRACT

One of the most challenging objectives of 3D cell culture is the development of scaffolding materials with outstanding biocompatibility and favorable mechanical strength. In this study, we fabricated a novel nanofibrous scaffold composed of fish collagen (FC) and polycaprolactone (PCL) blends by using the electrospinning method. Nanofibrous scaffolds were characterized using a scanning electron microscope (SEM), and it was revealed that the diameter of nanofibers decreased as FC content was increased in the FC/PCL composite nanofibers. The cytocompatibility of the FC/PCL scaffolds was evaluated by SEM, WST-1 assay, confocal microscopy, western blot, and RT-PCR. It was found that the scaffolds not only facilitated the adhesion, spreading, protrusions, and proliferation of thymic epithelial cells (TECs), but also stimulated the expression of genes and proteins involved in cell adhesion and T-cell development. Thus, these results suggest that the FC/PCL composite nanofibrous scaffolds will be a useful model of 3D cell culture for TECs and may have wide applicability in the future for engineering tissues or organs.


Subject(s)
Biocompatible Materials/chemistry , Cell Culture Techniques/methods , Collagen/chemistry , Fishes/metabolism , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Proliferation , Epithelial Cells/cytology , Mice , Nanofibers/chemistry , Thymus Gland/cytology , Tissue Engineering/methods
4.
Biochem Biophys Res Commun ; 454(1): 144-50, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25450371

ABSTRACT

Pancreatic adenocarcinoma up-regulated factor (PAUF) is expressed in pancreatic ductal adenocarcinoma (PDAC) and plays an important role in tumor progression and metastasis. Here we evaluate the anti-tumor efficacy of a human monoclonal antibody against PAUF, PMAb83, to provide a therapeutic intervention to treat the disease. PMAb83 reduced tumor growth and distant metastasis in orthotopically xenografted mice of human PDAC cells. PMAb83 treatment retarded proliferation along with weakened aggressiveness traits of the carcinoma cells. AKT/ß-catenin signaling played a role in the carcinoma cell proliferation and the treated xenograft tumors exhibited reduced levels of ß-catenin and cyclin D1. Moreover PMAb83 abrogated the PAUF-induced angiogenic responses of endothelial cells, reducing the density of CD31(+) vessels in the treated tumors. In combination with gemcitabine, PMAb83 conferred enhanced survival of xenografted mice by about twofold compared to gemcitabine alone. Taken together, our findings show that PMAb83 treatment decreases the aggressiveness of carcinoma cells and suppresses tumor vascularization, which culminates in mitigated tumor growth and metastasis with improved survival in PDAC mouse models.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Lectins/antagonists & inhibitors , Lectins/immunology , Pancreatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Combined Modality Therapy , Cyclin D1/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Progression , Endothelial Cells/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Nude , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , beta Catenin/metabolism , Gemcitabine
5.
Biochem Biophys Res Commun ; 446(1): 1-7, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24434139

ABSTRACT

TMPRSS4 is a novel type II transmembrane serine protease that is highly expressed on the cell surface in pancreatic, thyroid, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates cancer cell invasion, epithelial-mesenchymal transition, and metastasis and that increased TMPRSS4 expression correlates with colorectal cancer progression. We also demonstrated that TMPRSS4 upregulates urokinase-type plasminogen activator (uPA) gene expression to induce cancer cell invasion. However, it remains unknown how proteolytic activity of TMPRSS4 contributes to invasion. In this study, we report that TMPRSS4 directly converted inactive pro-uPA into the active form through its proteolytic activity. Analysis of conditioned medium from cells overexpressing TMPRSS4 demonstrated that the active TMPRSS4 protease domain is released from the cells and is associated with the plasma membrane. Furthermore, TMPRSS4 could increase pro-uPA-mediated invasion in a serine proteolytic activity-dependent manner. These observations suggest that TMPRSS4 is an upstream regulator of pro-uPA activation. This study provides valuable insights into the proteolytic function of TMPRSS4 as well as mechanisms for the control of invasion.


Subject(s)
Membrane Proteins/metabolism , Neoplasm Invasiveness/physiopathology , Serine Endopeptidases/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Enzyme Activation , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Up-Regulation , Urokinase-Type Plasminogen Activator/genetics
6.
Cell Signal ; 26(2): 398-408, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23978400

ABSTRACT

TMPRSS4 is a novel type II transmembrane serine protease that is highly expressed in pancreatic, thyroid, colon, and other cancer tissues. Previously, we demonstrated that TMPRSS4 mediates tumor cell invasion, migration, and metastasis. However, the mechanisms by which TMPRSS4 contributes to invasion are not fully understood. Here, we demonstrated that TMPRSS4 induced the transcription of the urokinase-type plasminogen activator (uPA) gene through activating the transcription factors Sp1, Sp3, and AP-1 in mainly a JNK-dependent manner and that the induction of uPA was required for TMPRSS4-mediated cancer cell invasion and signaling events. In addition, the uPA receptor was involved in TMPRSS4-induced signaling activation and subsequent uPA expression probably through its association with TMPRSS4 on the cell surface. Immunohistochemical analysis showed that uPA expression was significantly correlated with TMPRSS4 expression in human lung and prostate cancers. These observations suggest that TMPRSS4 is an important regulator of uPA gene expression; the upregulation of uPA by TMPRSS4 contributes to invasion and may represent a novel mechanism for the control of invasion.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Signal Transduction , Up-Regulation , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Cell Movement , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , RNA Interference , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Serine Endopeptidases/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/genetics
7.
Bioorg Med Chem Lett ; 23(6): 1748-51, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23414802

ABSTRACT

TMPRSS4 is a novel type II transmembrane serine protease that has been implicated in the invasion and metastasis of colon cancer cells. In this study, a novel series of 2-hydroxydiarylamide derivatives were synthesized and evaluated for inhibiting TMPRSS4 serine protease activity and suppressing cancer cell invasion. These derivatives demonstrated good inhibitory activity against TMPRSS4 serine protease, which correlated with the promising anti-invasive activity of colon cancer cells overexpressing TMPRSS4.


Subject(s)
Amides/chemistry , Membrane Proteins/antagonists & inhibitors , Serine Proteinase Inhibitors/chemistry , Amides/metabolism , Amides/toxicity , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Drug Evaluation, Preclinical , Humans , Membrane Proteins/metabolism , Protein Binding , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/metabolism , Serine Proteinase Inhibitors/toxicity , Structure-Activity Relationship
8.
Int J Mol Med ; 28(2): 255-60, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21567078

ABSTRACT

The hepatitis B virus X (HBX) protein, a regulatory protein of the hepatitis B virus (HBV), has been shown to generate reactive oxygen species (ROS) in human liver cell lines; however, the mechanism by which cells protect themselves under this oxidative stress is poorly understood. Here, we show that HBX induces the up-regulation of Forkhead box class O 4 (Foxo4) not only in Chang cells stably expressing HBX (Chang-HBX) but also in primary hepatic tissues from HBX-transgenic mice. HBX also increased ROS, but reduction of the abundance of ROS using N-acetylcystein (NAC) diminished the levels of Foxo4. Elevated Foxo4 was also detected in nuclei of Chang-HBX cells but not in Chang cells stably expressing the vector (Chang-Vec), suggesting that HBX activates the transcriptional activity of Foxo4. When we examined whether HBX bypasses JNK signaling that targets Foxo4, we found that the activity of JNK but not of ERK is required for the up-regulation of Foxo4 even in the presence of HBX. Furthermore, the reduction of Foxo4 levels using siRNA or a JNK inhibitor rendered Chang-HBX cells sensitive to apoptosis under oxidative stress, suggesting that up-regulation of Foxo4 mediated by HBX enhances resistances to oxidative stress-induced cell death. Accordingly, we propose that Foxo4 may be a useful target for suppression in the treatment of HBV-associated hepatocellular carcinoma cells.


Subject(s)
Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Oxidative Stress/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation , Animals , Cell Death , Cell Line , Cell Nucleus/metabolism , Forkhead Transcription Factors/antagonists & inhibitors , Gene Expression Regulation , Humans , MAP Kinase Kinase 4/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Transport/genetics , Reactive Oxygen Species/metabolism , Viral Regulatory and Accessory Proteins
9.
Exp Mol Med ; 43(5): 291-7, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21464589

ABSTRACT

Pancreatic cancer is a notorious disease with a poor prognosis and low survival rates, which is due to limited advances in understanding of the molecular mechanism and inadequate development of effective treatment options over the decades. In previous studies, we demonstrated that a novel soluble protein named pancreatic adenocarcinoma up-regulated factor (PAUF) acts on tumor and immune cells and plays an important role in metastasis and progression of pancreatic cancer. Here we show that PAUF promotes adhesiveness of pancreatic cancer cells to various extracellular matrix (ECM). Our results further support a positive correlation of activation and expression of focal adhesion kinase (FAK), a key player in tumor cell metastasis and survival, with PAUF expression. PAUF-mediated adhesiveness was significantly attenuated upon blockade of the FAK pathway. Moreover, PAUF appeared to enhance resistance of pancreatic cancer cells to anoikis via modulation of FAK. Our results suggest that PAUF-mediated FAK activation plays an important role in pancreatic cancer progression.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Lectins/metabolism , Pancreatic Neoplasms/metabolism , Anoikis/genetics , Cell Line, Tumor , Focal Adhesions/genetics , Humans , Intercellular Signaling Peptides and Proteins , Lectins/genetics , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/genetics
10.
Exp Mol Med ; 43(2): 82-90, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21196815

ABSTRACT

It is not yet understood how the enhanced expression of pancreatic adenocarcinoma up-regulated factor (PAUF; a novel oncogene identified in our recent studies), contributes to the oncogenesis of pancreatic cells. We herein report that PAUF up-regulates the expression and transcriptional activity of ß-catenin while the suppression of PAUF by shRNA down-regulates ß-catenin. The induction of b-catenin by PAUF is mediated by the activities of Akt and GSK-3ß, but inhibition of downstream ERK does not reduce ß-catenin expression. To test whether PAUF emulates either the Wnt3a-mediated or the protein kinase A-mediated signaling pathway for the stabilization of ß-catenin, we examined the phosphorylation status of ß-catenin in the presence of PAUF compared with that of ß-catenin during treatment with Wnt3a or dibutyryl cAMP, a cell permeable cyclic AMP analogue. PAUF expression induces phosphorylation at Ser-33/37/Thr-41 and Ser-675 of ß-catenin but no phosphorylation at Ser-45, indicating that a unique phosphorylation pattern of b-catenin is caused by PAUF. Finally, the expression of PAUF up-regulates both cyclin-D1 and c-Jun, target genes of ß-catenin, leading to a rapid proliferation of pancreatic cells; conversely decreased PAUF expression (by shRNA) results in the reduced proliferation of pancreatic cells. Treatment with hexachlorophene (an inhibitor of ß-catenin) reduces the proliferation of pancreatic cells despite the presence of PAUF. Taken together, we propose that PAUF can up-regulate and stabilize ß-catenin via a novel pattern of phosphorylation, thereby contributing to the rapid proliferation of pancreatic cancer cells.


Subject(s)
Adenocarcinoma , Lectins/metabolism , Pancreatic Neoplasms , Up-Regulation , beta Catenin/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins , Lectins/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , beta Catenin/genetics
11.
Int J Oncol ; 36(4): 1023-30, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198349

ABSTRACT

The tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has been used to treat a variety of cancer cells. However, since some gastric cancer cells are resistant to TRAIL, we explored whether reovirus induces cytolysis in TRAIL-resistant gastric cancer cells. We found that TRAIL-resistant SNU-216 gastric cancer cells were susceptible to apoptosis by reovirus infection. Furthermore, co-treatment with reovirus and TRAIL accelerated apoptosis of SNU-216 cells by down-regulation of Akt activation as assessed by a very low activation of Akt in TRAIL-sensitive SNU-668 gastric cancer cells. Inhibition of Akt signaling with wortmannin or suppression of Akt expression with sh-Akt lentivirus promoted reovirus-mediated apoptosis of SNU-216 gastric cancer cells. Reovirus infection also down-regulates the activation of signaling molecules such as Ras and ERK involved in cell proliferation and survival but not the activation of p38 MAPK involved in cellular stress. In addition, the co-treatment with reovirus and TRAIL resulted in cleavage of caspase-8, caspase-9 and Bid, leading to a decrease in the mitochondrial membrane potential, indicating that reovirus may utilize the mitochondrial intrinsic apoptotic pathway in TRAIL-resistant SNU-216 gastric cancer cells. Accordingly, we first demonstrate that reovirus infection down-regulates Akt activation, leading to apoptosis of TRAIL-resistant gastric cancer cells.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-akt/metabolism , Reoviridae/pathogenicity , Stomach Neoplasms/virology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Androstadienes/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Enzyme Activation , Humans , Membrane Potential, Mitochondrial , Mice , Mitochondria/enzymology , Mitochondria/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Wortmannin
12.
Int J Oncol ; 35(3): 617-24, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19639182

ABSTRACT

Reovirus functions as an oncolytic agent for many types of cancer including colon cancer. Although most studies have emphasized the role of activated Ras signaling in enhancing reoviral oncolysis in susceptible cells, we note that many colon cancers also display elevated beta-catenin. Thus, it is possible that enhanced beta-catenin may augment reoviral susceptibility in colon cancer cells. To explore this hypothesis, HEK293 cells were treated with the glycogen synthase kinase (GSK)-3beta inhibitor LiCl, thereby inducing beta-catenin, followed by reoviral infection. Co-administration with LiCl indeed enhanced cell death compared to reovirus infection alone, but this was not associated with elevated reoviral replication. Similarly, HEK293 cells expressing the Frizzled-1 receptor in Wnt3a-conditioned medium also showed reovirus replication equivalent to that in cells in control medium, further suggesting that up-regulation of beta-catenin does not enhance the replication of reovirus. Instead, we observed that inhibition of GSK-3beta with LiCl decreased reovirus-induced NF-kappaB activation, leading to accelerated apoptosis via caspase 8 activation. We further found that colon cancer HCT116 cells were sensitized to apoptosis by co-treatment with reovirus and a GSK-3beta inhibitor, AR-A014418. Finally, we identified that inhibition of NF-kappaB sensitized apoptosis of HEK293 or HCT 116 cells during reovirus infection. Taken together, we propose that inhibition of GSK-3beta sensitizes reovirus-induced apoptosis of colon cancer cells by down-regulation of NF-kappaB activity, offering a potentially improved therapeutic strategy for the treatment of colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/virology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Oncolytic Virotherapy/methods , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/therapy , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta , Humans , NF-kappa B/drug effects , NF-kappa B/metabolism , Orthoreovirus, Mammalian/physiology , Reoviridae Infections/virology , Signal Transduction/drug effects , Signal Transduction/physiology , beta Catenin/drug effects , beta Catenin/metabolism
13.
Cancer Lett ; 276(2): 136-42, 2009 Apr 18.
Article in English | MEDLINE | ID: mdl-19091460

ABSTRACT

Many studies have shown that the activation of beta-catenin signaling can promote oncogenesis, and it is therefore of interest to find agents that modulate this pathway. Recent work has shown using B lymphoma cells that infection by Epstein-Barr virus (EBV) and expression of its latent membrane protein (LMP)-1, cause increases in the expression of beta-catenin and cellular transformation. Conversely, results from cell-based small molecule screening studies have shown that the antibiotic hexachlorophene can down-regulate beta-catenin in colon cancer cells. Here we report that hexachlorophene also counteracts the elevated beta-catenin levels in EBV-infected B lymphomas. This is associated with restoration in levels of Siah-1 (an E3 ubiquitin ligase that is active in beta-catenin regulation) which had been diminished by LMP-1. Our results suggest that Siah-1 is targeted by both LMP-1 and hexachlorophene with opposite effects. The hexachlorophene modulation of Siah-1 and beta-catenin is independent of p53 and results in reduced expression of cyclin-D1 and c-Myc (target genes of beta-catenin), leading to the growth arrest of B lymphoma cells. From these results we propose that hexachlorophene may provide a novel therapeutic strategy for EBV-infected B lymphoma cells by reducing beta-catenin levels via the restoration of Siah-1.


Subject(s)
Herpesvirus 4, Human/physiology , Hexachlorophene/pharmacology , Lymphoma, B-Cell/drug therapy , Nuclear Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , beta Catenin/metabolism , Cell Line, Tumor , Humans , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/virology , Proteasome Endopeptidase Complex/physiology , Tumor Suppressor Protein p53/physiology , Up-Regulation , Viral Matrix Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...