Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
BMC Cancer ; 14: 324, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24885002

ABSTRACT

BACKGROUND: Squamous cell carcinoma of the oral cavity (OSCC) is a common cancer form with relatively low 5-year survival rates, due partially to late detection and lack of complementary molecular markers as targets for treatment. Molecular profiling of head and neck cancer has revealed biological similarities with basal-like breast and lung carcinoma. Recently, we showed that 16 genes were consistently altered in invasive breast tumors displaying varying degrees of aggressiveness. METHODS: To extend our findings from breast cancer to another cancer type with similar characteristics, we performed an integrative analysis of transcriptomic and proteomic data to evaluate the prognostic significance of the 16 putative breast cancer-related biomarkers in OSCC using independent microarray datasets and immunohistochemistry. Predictive models for disease-specific (DSS) and/or overall survival (OS) were calculated for each marker using Cox proportional hazards models. RESULTS: We found that CBX2, SCUBE2, and STK32B protein expression were associated with important clinicopathological features for OSCC (peritumoral inflammatory infiltration, metastatic spread to the cervical lymph nodes, and tumor size). Consequently, SCUBE2 and STK32B are involved in the hedgehog signaling pathway which plays a pivotal role in metastasis and angiogenesis in cancer. In addition, CNTNAP2 and S100A8 protein expression were correlated with DSS and OS, respectively. CONCLUSIONS: Taken together, these candidates and the hedgehog signaling pathway may be putative targets for drug development and clinical management of OSCC patients.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Mouth Neoplasms/chemistry , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Multivariate Analysis , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phenotype , Predictive Value of Tests , Proportional Hazards Models , Risk Factors , Time Factors , Tumor Burden , Young Adult
2.
Proteomics ; 11(3): 352-60, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21268265

ABSTRACT

Ovarian teratoma is a dermoid cyst in the ovary that contains mature tissues such as hair, teeth, bone, thyroid, etc. To understand the molecular mechanisms of ovarian teratoma growth, a comparative proteomic analysis was undertaken using mesenchymal stem cell-like cells (MSCLCs) isolated from normal human ovarian or teratoma tissues. Both normal ovarian and teratoma MSCLCs expressed stem cell markers OCT4 and NANOG, and were negatively staining with the senescence-associated (SA) ß-galactosidase. Furthermore, teratoma MSCLCs had higher proliferation and colony formation rates, with more angiogenic property than that of normal MSCLCs. Proteomic study revealed that 17 proteins had the expression changes over eightfold in ovarian teratoma MSCLCs compared with normal control. Interestingly, among them, GSTM2 was strongly expressed in teratoma MSCLCs. Moreover, overexpressed GSTM2 in the teratoma was associated with downregulation of p38 MAPK and activation of AKT and survivin. Taken together, these findings suggest that that ovarian teratoma MSCLCs have a higher potency for proliferation and angiogenesis and GSTM2 appears to be involved in the regulation of other survival genes.


Subject(s)
Biomarkers, Tumor/metabolism , Glutathione Transferase/metabolism , Mesenchymal Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Proteome/analysis , Teratoma/metabolism , Adult , Animals , Apoptosis , Blotting, Western , Cell Movement , Cell Proliferation , Cells, Cultured , Dermoid Cyst/metabolism , Dermoid Cyst/pathology , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunoenzyme Techniques , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Teratoma/pathology
3.
J Cell Biochem ; 112(6): 1552-62, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21344490

ABSTRACT

It has drawn a lot of attention to target signal transducer and activator of transcription 3 (STAT3) as a potential strategy for cancer therapeutics. Using several myelogenous cell lines, the effect of genipin (an active compound of Gardenia fruit) on the STAT3 pathway and apoptosis was investigated. Genipin suppressed the constitutive STAT3 activation in U266 and U937 cells and stimulated Src homology 2 domain-containing phosphatase 1 (SHP-1), which dephosphorylates and inactivates STAT3. Specifically, genipin blocked STAT3 activation via repressing the activation of c-Src, but not Janus kinase 1 (JAK1). Genipin also downregulated the expression of STAT3 target genes including Bcl-2, Bcl-x(L) , Survivin, Cyclin D1, and VEGF. Conversely, protein tyrosine phosphatase inhibitor pervanadate blocked genipin induced STAT3 inactivation. Using DNA fragmentation or TUNEL assays, we demonstrated the apoptotic effect of genipin on U266, MM.1S, and U937 cells. Furthermore, genipin effectively potentiated the cytotoxic effect of chemotherapeutic agents, such as bortezomib, thalidomide, and paclitaxel in U266 cells. Our data suggest that through regulation of Src and SHP-1, genipin antagonizes STAT3 for the induction of apoptosis in myeloma cells.


Subject(s)
Apoptosis/drug effects , Multiple Myeloma/metabolism , STAT3 Transcription Factor/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Humans , In Situ Nick-End Labeling , Iridoid Glycosides , Iridoids , Multiple Myeloma/genetics , Pyrazines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
4.
Breast Cancer Res ; 13(6): R113, 2011.
Article in English | MEDLINE | ID: mdl-22085699

ABSTRACT

INTRODUCTION: Tobacco smoke is known to be the main cause of lung, head and neck tumors. Recently, evidence for an increasing breast cancer risk associated with tobacco smoke exposure has been emerging. We and other groups have shown that nicotine, as a non-conventional carcinogen, has the potential to facilitate cancer genesis and progression. However, the underlying mechanisms by which the smoke affects the breast, rather than the lung, remain unclear. Here, we examine possible downstream signaling pathways of the nicotinic acetylcholine receptor (nAChR) and their role in breast cancer promotion. METHODS: Using human benign MCF10A and malignant MDA-MB-231 breast cells and specific inhibitors of possible downstream kinases, we identified nAChR effectors that were activated by treatment with nicotine. We further tested the effects of these effector pathways on the regulation of E2F1 activation, cell cycle progression and on Bcl-2 expression and long-term cell survival. RESULTS: In this study, we demonstrated a novel signaling mechanism by which nicotine exposure activated Src to sensitize epidermal growth factor receptor (EGFR)-mediated pathways for breast cancer cell growth promotion. After the ligation of nAChR with nicotine, EGFR was shown to be activated and then internalized in both MCF10A and MDA-MB-231 breast cancer cells. Subsequently, Src, Akt and ERK1/2 were phosphorylated at different time points following nicotine treatment. We further demonstrated that through Src, the ligation of nicotine with nAChR stimulated the EGFR/ERK1/2 pathway for the activation of E2F1 and further cell progression. Our data also showed that Akt functioned directly downstream of Src and was responsible for the increase of Bcl-2 expression and long-term cell survival. CONCLUSIONS: Our study reveals the existence of a potential, regulatory network governed by the interaction of nicotine and nAChR that integrates the conventional, mitogenic Src and EGFR signals for breast cancer development.


Subject(s)
Breast Neoplasms/metabolism , Carcinogens/pharmacology , ErbB Receptors/metabolism , Nicotine/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , E2F1 Transcription Factor/metabolism , ErbB Receptors/agonists , Female , Humans , MAP Kinase Signaling System/drug effects , Protein Binding , Receptors, Nicotinic/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , src-Family Kinases/metabolism
5.
Apoptosis ; 16(7): 696-707, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21519916

ABSTRACT

Cryptotanshinone is a biologically active compound from the root of Salvia miltiorrhiza. In the present study, we investigated the molecular mechanisms by which cryptotanshinone is in synergy with tumor necrosis factor-alpha (TNF-α) for the induction of apoptosis in human chronic myeloid leukemia (CML) KBM-5 cells. The co-treatment of cryptotanshinone with TNF-α reduced the viability of the cells [combination index (CI) < 1]. Concomitantly, the co-treatment of cryptotanshinone and TNF-α elicited apoptosis, manifested by enhanced the number of terminal deoxynucleotide transferase-mediated dUTP-nick-end labeling (TUNEL)-positive cells, the sub-G1 cell populations, and the activation of caspase-8 and -3, in comparison with the treatment with either drug alone. The treatment with cryptotanshinone further suppressed TNF-α-mediated expression of c-FLIP(L), Bcl-x(L), but the increased level of tBid (a caspase-8 substrate). Furthermore, cryptotanshinone activated p38 but not NF-κB in TNF-α-treated KBM-5 cells. The addition of a specific p38 MAPK inhibitor SB203580 significantly attenuated cryptotanshinone/TNF-α-induced apoptosis. The combination treatment of cryptotanshinone and TNF-α also stimulated the reactive oxygen species (ROS) generation. N-acetyl-L-cysteine (NAC, a ROS scavenger) was not only able to block cryptotanshinone/TNF-α-induced ROS production but also the activation of caspase-8 and p38 MAPK. Overall, our findings suggest that cryptotanshinone can sensitize TNF-α-induced apoptosis in human myeloid leukemia KBM-5 cells, which appears through ROS-dependent activation of caspase-8 and p38.


Subject(s)
Apoptosis/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Phenanthrenes/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Enzyme Activation/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Phenanthrenes/chemistry , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
6.
J Pineal Res ; 51(1): 87-93, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21392092

ABSTRACT

Sphingosine kinase 1 (SPHK1) is a newly discovered modulator of hypoxia inducible factor 1α (HIF-1α) with various biological activities such as cell growth, survival, invasion, angiogenesis, and carcinogenesis. Thus, in the present study, the biological mechanisms of melatonin were elucidated in association with SPHK1 pathway in PC-3 prostate cancer cells under hypoxia. Melatonin inhibited the stability of HIF-1α in a time- and concentration- dependent manners. Also, melatonin decreased SPHK1 activity in PC-3 cells during hypoxia. Furthermore, melatonin suppressed AKT/glycogen synthase kinase-3ß (GSK-3ß) signaling pathway, which stabilizes HIF-1α via inhibition of von Hippel-Lindau tumor suppressor protein. Consistently, siRNA-SPHK1 and sphingosine kinase inhibitor (SKI) effectively blocked the expression of HIF-1α, phospho-AKT and vascular endothelial growth factor (VEGF) production in PC-3 cells under hypoxia, suggesting the role of SPHK1 in melatonin-inhibited HIF-1α accumulation. Moreover, reactive oxygen species (ROS) scavenger N-acteylcysteine enhanced melatonin-inhibited HIF-1α expression and SPHK1 activity. Overall, our findings suggest that melatonin suppresses HIF-1α accumulation via inhibition of SPHK1 pathway and ROS generation in PC-3 cells under hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melatonin/pharmacology , Metabolic Networks and Pathways/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prostatic Neoplasms/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Statistics, Nonparametric , Vascular Endothelial Growth Factor A/metabolism
7.
Biol Pharm Bull ; 34(6): 850-5, 2011.
Article in English | MEDLINE | ID: mdl-21628883

ABSTRACT

Hypoxia inducible factor 1 alpha (HIF-1α) is frequently over-expressed in the numerous types of cancer and plays an important role in angiogenesis. In the present study, the inhibitory mechanism of rhapontigenin isolated from Vitis coignetiae was investigated on HIF-1α stability and angiogenesis in human prostate cancer PC-3 cells. Rhapontigenin significantly suppressed HIF-1α accumulation at protein level but not at mRNA level in PC-3 cells under hypoxia. Also, rhapontigenin suppressed hypoxia-induced HIF-1α activation in various cancer cells, such as colorectal adenocarcinoma (SW620), breast adenocarcinoma (MCF-7), fibrosarcoma (HT-1080) and prostate carcinoma (LNCaP). Interestingly, rhapontigenin had more potency in inhibition of hypoxia-induced HIF-1α expression than that of resveratrol, a known HIF-1α inhibitor. In addition, rhapontigenin promoted hypoxia-induced HIF-1α degradation and cycloheximide (CHX) blocked protein synthesis. A prolyl hydroxylase (PHD) inhibitor dimethyloxalylglycine (DMOG) is usually utilized to examine whether prolyl hydroxylation is involved in inhibition of HIF-1α accumulation. Here, DMOG recovered HIF-1α accumulation inhibited by rhapontigenin. Immunoprecipitation assay also revealed that rhapotigenin enhanced the binding of hydroxylated HIF-1α to von Hippel-Lindau (VHL) tumor suppressor protein. Furthermore, rhapontigenin reduced vascular endothelial growth factor (VEGF) secretion in hypoxic PC-3 cells as well as suppressed tube formation in human umbilical vein endothelial cells (HUVECs) treated by the conditioned media of hypoxic PC-3 cells. However, anti-angiogenic effect of rhapontigenin in hypoxic PC-3 cells was reversed by DMOG. Taken together, these findings suggest that rhapontigenin inhibits HIF-1α accumulation and angiogenesis in PC-3 prostate cancer cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma/drug therapy , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/prevention & control , Prostatic Neoplasms/drug therapy , Stilbenes/pharmacology , Amino Acids, Dicarboxylic/pharmacology , Angiogenesis Inhibitors/antagonists & inhibitors , Carcinoma/metabolism , Cell Line , Cell Line, Tumor , Culture Media, Conditioned/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Female , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Neoplasms/drug therapy , Neoplasms/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational/drug effects , Stilbenes/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
8.
Biol Pharm Bull ; 34(1): 41-6, 2011.
Article in English | MEDLINE | ID: mdl-21212515

ABSTRACT

Anethole is known to possess anti-inflammatory and anti-tumor activities and to be a main constituent of fennel, anise, and camphor. In the present study, we evaluated anti-metastatic and apoptotic effects of anethole on highly-metastatic HT-1080 human fibrosarcoma tumor cells. Despite weak cytotoxicity against HT-1080 cells, anethole inhibited the adhesion to Matrigel and invasion of HT-1080 cells in a dose-dependent manner. Anethole was also able to down-regulate the expression of matrix metalloproteinase (MMP)-2 and -9 and up-regulate the gene expression of tissue inhibitor of metalloproteinase (TIMP)-1. The similar inhibitory effect of anethole on MMP-2 and -9 activities was confirmed by zymography assay. Furthermore, anethole significantly decreased mRNA expression of urokinase plasminogen activator (uPA), but not uPA receptor (uPAR). In addition, anethole suppressed the phosphorylation of AKT, extracellular signal-regulated kinase (ERK), p38 and nuclear transcription factor kappa B (NF-κB) in HT-1080 cells. Taken together, our findings indicate that anethole is a potent anti-metastatic drug that functions through inhibiting MMP-2/9 and AKT/mitogen-activated protein kinase (MAPK)/NF-κB signal transducers.


Subject(s)
Anisoles/pharmacology , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Allylbenzene Derivatives , Animals , Anisoles/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Molecular Structure , Neoplasm Metastasis/prevention & control , Signal Transduction
9.
J Biol Chem ; 284(39): 26315-21, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19648117

ABSTRACT

The retinoblastoma protein (Rb) plays a pivotal role in regulating cell proliferation and apoptosis. Nutlin-3, a small molecule MDM2 antagonist blocking interaction between MDM2 and p53, activates p53 resulting in cell growth arrest or apoptosis in various cancer cells. However, the molecular basis for the different cellular responses upon nutlin-3 treatment is not fully understood. In this study, we show that nutlin-3 activates p53 resulting in a dramatic increase in MDM2 expression and a marked reduction in total Rb protein levels. Interestingly, nutlin-3 reduces the levels of hypophosphorylated Rb and induces massive apoptosis in SJSA-1 cells, which can be largely rescued by knockdown of MDM2 or by expression of constitutively active Rb. By contrast, nutlin-3 treatment of several human cancer cells, including A549, U2-OS, and HCT116, results in an accumulation of hypophosphorylated Rb and cell cycle arrest but not apoptosis. Furthermore, we show that down-regulation of Rb by nutlin-3 does not lead to E2F1 activation nor does E2F1 play a critical role for nutlin-3-induced apoptosis in SJSA-1 cells. Taken together, these results suggest that Rb plays a critical role in influencing cellular response to activation of p53 pathway by nutlin-3.


Subject(s)
Imidazoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Flow Cytometry , HCT116 Cells , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Signal Transduction/drug effects
10.
J Cell Biochem ; 109(1): 152-61, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19911375

ABSTRACT

Cigarette smoking is a mixture of thousands of compounds, many of which are carcinogens, such as NNK [4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone]. Nicotine, as an addictive substance in cigarette, has been shown to promote growth of non-neuronal cells. It is unclear how nicotine cooperates with tobacco-related carcinogens during tumorigenesis. Here, by concurrent treatment of nicotine and NNK, we investigate the effect of the cooperation of these two compounds on cell growth and apoptosis in various different lung epithelial (RLE) or cancer (LKR) cells. We demonstrated that short-term nicotine exposure moderately activated mitogenic signaling pathways (such as PKC, ERK, and Akt) and a mediocre protection against cisplatin-mediated apoptosis. In contrast, NNK strongly stimulated mitogenic signaling and rendered the cells a high resistance to cisplatin. The pre-ligation of nAChR by nicotine interfered with NNK-mediated mitogenic signaling and resistance to cisplatin, the magnitude of which was similar as that exposed to nicotine alone. Interestingly, a week after the exposure to nicotine or nicotine plus NNK, Bcl-2 expression was augmented, accompanied with the increased resistance to cisplatin-induced apoptosis. In comparison, long-term NNK treatment provided very little protection of the cells from cisplatin. We also showed that the combination treatment promoted more cells to grow in an anchorage-independent fashion than NNK exposure alone. Thus, the data suggest that through occupying nAChR, nicotine appears to modulate NNK-mediated signaling and persistently sustain pro-survival activities to promote transformation.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Nitrosamines/toxicity , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , DNA Fragmentation , Humans , Immunoblotting , Mice , Rats , Receptors, Nicotinic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation
11.
Proteome Sci ; 8: 67, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21159204

ABSTRACT

BACKGROUND: Sulforaphane (SFN) is an isothiocyanate found in cruciferous vegetables that exerts anti-oxidant, anti-inflammatory, anti-cancer and radio-sensitizing activities. Nonetheless, the mechanism responsible for SFN-induced cell death is not fully understood. In the present study, anti-cancer mechanism of SFN was elucidated in LNCaP prostate cancer cells. RESULTS: SFN exerted cytotoxicity and increased TUNEL positive cells in a concentration-dependent manner in LNCaP cells. Proteomics study revealed that levels of nine proteins including tubulin ß-2, phosphoglucomutase-3 (PGM3), melanoma-derived leucine zipper containing extra-nuclear factor, activin A type I receptor precursor, smoothelin-A, KIA0073, hypothetical protein LOC57691 and two unnamed proteins were changed over 8 folds in SFN treated LNCaP cells compared to untreated control. We have further confirmed that SFN reduced PGM3 expression with western blotting and showed that PGM3 siRNA enhanced cytotoxicity demonstrated by cell morphology and TUNEL assays in LNCaP cells. CONCLUSION: Taken together, these findings suggest that PGM3 plays a role in mediating SFN-induced cell death in LNCaP cells, and is a potential molecular therapeutic target for prostate cancer.

12.
J Cell Biochem ; 107(1): 76-85, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19241442

ABSTRACT

It is known that Ras mutations, together with loss of PKC, are apoptotic in various types of mammalian cells. The mechanism of how aberrant Ras transmits this apoptotic signaling remains unclear. Using three V12-Ha-ras loop mutants that preferentially bind to and activate one of Ras effectors, we tested the role of Ras downstream pathways in the induction of apoptosis in rat lung epithelia, human lung or prostate cancer cells. After PKC inhibition, the activation of PI3K/Akt renders the susceptibility of cells to apoptosis. We also demonstrate that the amount of ROS is moderately increased in the cells ectopically expressing V12C40 and dramatically elevated by suppression of PKC, which leads to apoptosis through the activation of UPR. Thus, our study suggests that after PKC abrogation, PI3K functions downstream of Ras to perturb the state of cellular redox and signals to ER stress-regulated apoptotic machinery.


Subject(s)
Apoptosis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Cell Line , DNA Fragmentation , Flow Cytometry , Genes, ras , Humans , Immunoblotting , Male , Oxidation-Reduction , Oxidative Stress/physiology , Rats , Reactive Oxygen Species/metabolism
13.
Curr Med Chem ; 15(13): 1330-5, 2008.
Article in English | MEDLINE | ID: mdl-18537612

ABSTRACT

Phellinus Linteus (Berkeley & M. A. Curtis) Teng (PL) is a medicinal mushroom that has been practiced in oriental countries for centuries to prevent ailments as diverse as gastroenteric dysfunction, diarrhea, haemorrhage and cancers. In an effort to translate the Asian traditional medicines into western-accepted therapies, scientists have demonstrated that the extracts from fruit-bodies or mycelium of PL not only stimulate the hormonal and cell-mediated immune function and quench the inflammatory reactions caused by a variety of stimuli, but also suppress the tumor growth and metastasis. Mounting evidence from different research groups has shown that PL induces apoptosis in a host of murine and human carcinomas without causing any measurable toxic effects to their normal counterparts. Recently, research has been focused on the anti-tumor effect of PL, and in particular, on its ability to enhance some conventional chemotherapeutic drugs. These studies suggest PL to be a promising candidate as an alternative anticancer agent or a synergizer for existing antitumor drugs. Hereinafter, we summarize the present progress in elucidating the mechanisms underlying the potency of PL and its anti-tumor function. The fractionation and identification of the biologically active components from PL are also briefly introduced.


Subject(s)
Agaricales/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , Agaricales/immunology , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/toxicity , Humans , Phellinus , Plant Extracts , Polysaccharides/immunology , Polysaccharides/toxicity , Safety
14.
Mol Cell Biol ; 22(9): 2928-38, 2002 May.
Article in English | MEDLINE | ID: mdl-11940651

ABSTRACT

The tumor suppressor p53 eliminates cancer-prone cells via multiple mechanisms, including apoptosis. Ras elicits apoptosis in cells after protein kinase C (PKC) downregulation. However, the role of p53 in Ras-mediated apoptosis has not been fully investigated. Here, we demonstrate that mouse fibroblasts that express wild-type p53 are more susceptible to apoptosis elicited by PKC inhibition if Ras is transiently expressed or upregulated as opposed to stably expressed. In the latter case, p53 is frequently mutated. Transiently increased Ras activity induces Bax, and PKC inhibition augments this induction. Overexpression of E6 inactivates p53 and thereby suppresses both Bax induction and apoptosis. In contrast, Bax is not induced in stable ras transfectants, regardless of PKC inhibition. The data suggest that short- and long-term activation of Ras use a different mechanism(s) to initiate apoptosis. The status of p53 may contribute to such differences.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Survival , Fibroblasts , Flow Cytometry , Gene Expression Regulation, Neoplastic , Mice , Mutation , Oligonucleotides, Antisense , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
15.
Cell Signal ; 14(10): 849-59, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12135706

ABSTRACT

Ras plays an important role in T cell signal transduction through multiple pathways. Here, we demonstrate that, upon stimulation, increasing Ras activity partially substitutes for calcium-mediated signals leading to IL-2 induction. The increase of Ras activity renders Jurkat cells the resistant to cyclosporin A (CsA) through increasing calcineurin activity. Coincidentally, the inducible binding of NIL-2 to a negative-regulatory element in the IL-2 promoter becomes less sensitive to CsA in the cells with increasing Ras activity. The dose of CsA required for inhibition of IL-2 induction in the cells with increased Ras activity remains similar to the concentration of CsA needed for the suppression of NFAT activation in control cells. The results suggest that Ras regulates calcium/calcineurin signalling during T cell activation and the existence of new immune-related target(s) for CsA action at the posttranscriptional level.


Subject(s)
Calcium Signaling/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Nuclear Proteins , T-Lymphocytes/immunology , Up-Regulation/immunology , ras Proteins/immunology , Calcineurin/drug effects , Calcineurin/metabolism , Calcium Signaling/drug effects , Cyclosporine/pharmacology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Genes, Regulator/drug effects , Genes, Regulator/immunology , Humans , Immunosuppressive Agents/pharmacology , Interleukin-2/genetics , Interleukin-2/immunology , Jurkat Cells , Lymphocyte Activation/drug effects , NFATC Transcription Factors , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/immunology , Protein Binding/drug effects , Protein Binding/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transcription Factors/drug effects , Transcription Factors/immunology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Up-Regulation/drug effects , ras Proteins/drug effects , ras Proteins/metabolism
16.
Oncotarget ; 6(31): 30939-56, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26439985

ABSTRACT

The synthetic curcumin analog B5 is a potent inhibitor of thioredoxin reductase (TrxR) that has potential anticancer effects. The molecular mechanism underlying B5 as an anticancer agent is not yet fully understood. In this study, we report that B5 induces apoptosis in two human cervical cancer cell lines, CaSki and SiHa, as evidenced by the downregulation of XIAP, activation of caspases and cleavage of PARP. The involvement of the mitochondrial pathway in B5-induced apoptosis was suggested by the dissipation of mitochondrial membrane potential and increased expression of pro-apoptotic Bcl-2 family proteins. In B5-treated cells, TrxR activity was markedly inhibited with concomitant accumulation of oxidized thioredoxin, increased formation of reactive oxygen species (ROS), and activation of ASK1 and its downstream regulatory target p38/JNK. B5-induced apoptosis was significantly inhibited in the presence of N-acetyl-l-cysteine. Microscopic examination of B5-treated cells revealed increased presence of cytoplasmic vacuoles. The ability of B5 to activate autophagy in cells was subsequently confirmed by cell staining with acridine orange, accumulation of LC3-II, and measurement of autophagic flux. Unlike B5-induced apoptosis, autophagy induced by B5 is not ROS-mediated but a role for the AKT and AMPK signaling pathways is implied. In SiHa cells but not CaSki cells, B5-induced apoptosis was promoted by autophagy. These data suggest that the anticarcinogenic effects of B5 is mediated by complex interplay between cellular mechanisms governing redox homeostasis, apoptosis and autophagy.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Curcumin/analogs & derivatives , Curcumin/pharmacology , Mitochondria/pathology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Uterine Cervical Neoplasms/pathology , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism
17.
PLoS One ; 7(7): e40435, 2012.
Article in English | MEDLINE | ID: mdl-22848379

ABSTRACT

Pancreatic cancer is a devastating human malignancy and gain of functional mutations in K-ras oncogene is observed in 75%-90% of the patients. Studies have shown that oncogenic ras is not only able to promote cell growth or survival, but also apoptosis, depending upon circumstances. Using pancreatic cancer cell lines with or without expressing mutated K-ras, we demonstrated that the inhibition of endogenous PKC activity sensitized human pancreatic cancer cells (MIA and PANC-1) expressing mutated K-ras to apoptosis, which had no apoptotic effect on BxPC-3 pancreatic cancer cells that contain a normal Ras as well as human lung epithelial BAES-2B cells. In this apoptotic process, the level of ROS was increased and PUMA was upregulated in a p73-dependent fashion in MIA and PANC-1 cells. Subsequently, caspase-3 was cleaved. A full induction of apoptosis required the activation of both ROS- and p73-mediated pathways. The data suggest that PKC is a crucial factor that copes with aberrant K-ras to maintain the homeostasis of the pancreatic cancer cells harboring mutated K-ras. However, the suppression or loss of PKC disrupts the balance and initiates an apoptotic crisis, in which ROS and p73 appear the potential, key targets.


Subject(s)
Apoptosis , Mutation , Oncogene Protein p21(ras)/metabolism , Pancreatic Neoplasms/metabolism , Protein Kinase C/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Protein p21(ras)/genetics , Pancreatic Neoplasms/pathology , Protein Kinase C/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
PLoS One ; 7(4): e28706, 2012.
Article in English | MEDLINE | ID: mdl-22493659

ABSTRACT

BACKGROUND: The aim of this study is to determine anti-cancer effect of Icariside II purified from the root of Epimedium koreanum Nakai on human acute myeloid leukemia (AML) cell line U937. METHODOLOGY/PRINCIPAL FINDINGS: Icariside II blocked the growth U937 cells in a dose- and time-dependent manner. In this anti-proliferation process, this herb compound rendered the cells susceptible to apoptosis, manifested by enhanced accumulation of sub-G1 cell population and increased the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells. Icariside II was able to activate caspase-3 and cleaved poly (ADP-ribose) polymerase (PARP) in a time-dependent manner. Concurrently, the anti-apoptotic proteins, such as bcl-x(L) and survivin in U937 cells, were downregulated by Icariside II. In addition, Icariside II could inhibit STAT3 phosphorylation and function and subsequently suppress the activation of Janus activated kinase 2 (JAK2), the upstream activators of STAT3, in a dose- and time-dependent manner. Icariside II also enhanced the expression of protein tyrosine phosphatase (PTP) SH2 domain-containing phosphatase (SHP)-1, and the addition of sodium pervanadate (a PTP inhibitor) prevented Icariside II-induced apoptosis as well as STAT3 inactivation in STAT3 positive U937 cells. Furthermore, silencing SHP-1 using its specific siRNA significantly blocked STAT3 inactivation and apoptosis induced by Icariside II in U937 cells. CONCLUSIONS/SIGNIFICANCE: Our results demonstrated that via targeting STAT3-related signaling, Icariside II sensitizes U937 cells to apoptosis and perhaps serves as a potent chemotherapeutic agent for AML.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Epimedium/chemistry , Flavonoids/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Plant Roots/chemistry , STAT3 Transcription Factor/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flavonoids/isolation & purification , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/genetics , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Leukemia, Myeloid, Acute/pathology , Phosphorylation , Plant Extracts/chemistry , Poly(ADP-ribose) Polymerases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Survivin , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics
19.
Cancer Lett ; 319(2): 197-202, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22261340

ABSTRACT

We investigated the molecular mechanisms responsible for fisetin-induced apoptosis in U266 cells. Fisetin elicited the cytotoxicity in U266 cells, manifested as an increased fraction of the cells with sub-G1 content or stained positively with TUNEL labeling. Fisetin enhanced caspase-3 activation, downregulation of Bcl-2 and Mcl-1(L), and upregulation of Bax, Bim and Bad. Fisetin activated AMPK as well as its substrate acetyl-CoA carboxylase (ACC), along with a decreased phosphorylation of AKT and mTOR. Fisetin also stimulated generation of ROS in U266 cells. Conversely, compound C or N-acetyl-L-cystein blocked fisetin-induced apoptosis. Our data suggest that fisetin-induced apoptosis in U266 cells is through ROS and AMPK pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Flavonoids/pharmacology , Multiple Myeloma/metabolism , Reactive Oxygen Species/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Flavonols , Humans , Signal Transduction/drug effects
20.
PLoS One ; 6(4): e18619, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21559516

ABSTRACT

As an addictive substance, nicotine has been suggested to facilitate pro-survival activities (such as anchorage-independent growth or angiogenesis) and the establishment of drug resistance to anticancer therapy. Tobacco smoking consists of a variety of carcinogens [such as benzopyrene (BP) and nitrosamine derivatives] that are able to cause DNA double strand breaks. However, the effect of nicotine on DNA damage-induced checkpoint response induced by genotoxins remains unknown. In this study, we investigated the events occurred during G(1) arrest induced by γ-radiation or BP in nicotine-treated murine or human lung epithelial cells. DNA synthesis was rapidly inhibited after exposure to γ-radiation or BP treatment, accompanied with the activation of DNA damage checkpoint. When these cells were co-treated with nicotine, the growth restriction was compromised, manifested by upregulation of cyclin D and A, and attenuation of Chk2 phosphorylation. Knockdown of cyclin D or Chk2 by the siRNAs blocked nicotine-mediated effect on DNA damage checkpoint activation. However, nicotine treatment appeared to play no role in nocodazole-induced mitotic checkpoint activation. Overall, our study presented a novel observation, in which nicotine is able to override DNA damage checkpoint activated by tobacco-related carcinogen BP or γ-irradiation. The results not only indicates the potentially important role of nicotine in facilitating the establishment of genetic instability to promote lung tumorigenesis, but also warrants a dismal prognosis for cancer patients who are smokers, heavily exposed second-hand smokers or nicotine users.


Subject(s)
DNA Damage/drug effects , Lung/drug effects , Nicotine/pharmacology , Animals , Benzopyrenes/pharmacology , Carcinogens , Cyclin A/biosynthesis , Cyclin D1/biosynthesis , Epithelial Cells/cytology , G1 Phase , Gamma Rays , Ganglionic Stimulants/pharmacology , Humans , Mice , Mutation , Nocodazole/pharmacology , Phosphorylation , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL