Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 38(4): 963-71, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21271218

ABSTRACT

Targeted therapy with reduced side effects is a major goal in cancer research. We investigated the effects of JS-K, a nitric oxide (NO) prodrug designed to release high levels of NO when suitably activated, on human breast cancer cell lines, on non-transformed human MCF-10A mammary cells, and on normal human mammary epithelial cells (HMECs). Cell viability assay, flow cytometry, electron microscopy, and Western blot analysis were used to study the effects of JS-K on breast cancer and on mammary epithelial cells. After a 3-day incubation, the IC50s of JS-K against the breast cancer cells ranged from 0.8 to 3 µM. However, JS-K decreased the viability of the MCF-10A cells by only 20% at 10-µM concentration, and HMECs were unaffected by 10 µM JS-K. Flow cytometry indicated that JS-K increased the percentages of breast cancer cells under-going apoptosis. Interestingly, flow cytometry indicated that JS-K increased acidic vesicle organelle formation in breast cancer cells, suggesting that JS-K induced autophagy in breast cancer cells. Electron microscopy confirmed that JS-K-treated breast cancer cells underwent autophagic cell death. Western blot analysis showed that JS-K induced the expression of microtubule light chain 3-II, another autophagy marker, in breast cancer cells. However, JS-K did not induce apoptosis or autophagy in normal human mammary epithelial cells. These data indicate that JS-K selectively induces programmed cell death in breast cancer cells while sparing normal mammary epithelial cells under the same conditions. The selective anti-tumor activity of JS-K warrants its further investigation in breast tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azo Compounds/pharmacology , Breast Neoplasms/pathology , Mammary Glands, Human/drug effects , Nitric Oxide/metabolism , Piperazines/pharmacology , Prodrugs/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Heme Oxygenase-1/metabolism , Humans , Mammary Glands, Human/pathology , Microtubule-Associated Proteins/metabolism , Up-Regulation
2.
Cancer ; 116(11 Suppl): 2806-14, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20503412

ABSTRACT

BACKGROUND: Although inflammatory breast cancer (IBC) is recognized as the most lethal variant of locally advanced breast cancer, few molecular signatures of IBC have been identified that can be used as targets to develop therapeutics that effectively inhibit the aggressive phenotype displayed by IBC tumors. METHODS: Real-time polymerase chain reaction analysis, Western blot analysis, modified Boyden chamber invasion assays, vasculogenic mimicry (VM) assays, and gelatin zymography were used in the current studies. Agonists and antagonists of the prostanoid receptors EP3 and EP4 and of EP4 short-hairpin RNA (shRNA) knockdown approaches were used as tools to assess the role of prostanoid receptors EP3 and EP4 in the regulation of specific biologic activities of IBC cells. RESULTS: The current studies revealed that the IBC breast cancer cell lines SUM149 and SUM190 express high levels of cyclooxygenase-2 messenger RNA and protein, produce abundant levels of prostaglandin E(2), and produce both EP3 and EP4 receptor proteins. Studies using the EP4 antagonist GW627368X and shRNA molecular knockdown approaches revealed a role for EP4 in regulating invasion of IBC cells. EP3, but not EP4, regulated the ability of SUM149 cells to undergo VM, which is the ability to form capillary-like structures, a characteristic exhibited by very aggressive tumor types. Inhibition of VM by sulprostone was associated with an inhibition of matrix metalloprotease-2 (MMP-2) enzyme activity. CONCLUSIONS: The prostanoid receptors EP3 and EP4 differentially regulate activities exhibited by IBC cells that have been associated with the aggressive phenotype of this lethal variant of breast cancer. Whereas EP4 regulates invasion, EP3 regulates VM and the associated increased MMP-2 enzyme activity.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Prostaglandin E/physiology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/metabolism , Isoindoles/pharmacology , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic , Prostaglandins E/metabolism , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype , Sulfonamides/pharmacology
3.
Biochem J ; 428(3): 429-37, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20388118

ABSTRACT

Signalling through the IFNalphaR (interferon-alpha receptor) and TCR (T-cell receptor) in Jurkat T lymphocytes results in distinct immune responses. Despite this both receptors elicit ERK (extracellular-signal-regulated kinase)/MAPK (mitogen-activated protein kinase) phosphorylation. Vav and Slp76 are shown to be required for IFNalpha (interferon-alpha)-stimulated ERK activity. These form a subset of proteins which behave identically on stimulation of both receptors. TCR deletion abrogates IFNalphaR-stimulated MAPK activity, whereas the canonical JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway is unaffected. Thus recruitment of the intact TCR ESC (early signalling complex) is necessary for this downstream MAPK response. Despite using a common ESC, stimulation of the IFNalphaR does not produce the transcriptional response associated with TCR. Up-regulation of the MAPK pathway by IFNalphaR might be important to ensure that the cell responds to only one stimulant.


Subject(s)
Receptor, Interferon alpha-beta/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/enzymology , Adaptor Proteins, Signal Transducing/metabolism , Humans , Jurkat Cells , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , T-Lymphocytes/immunology , Transfection
4.
Clin Exp Metastasis ; 26(3): 197-204, 2009.
Article in English | MEDLINE | ID: mdl-19112600

ABSTRACT

In breast tumors, high levels of leptin have been associated with increased incidence of breast cancer metastasis. Breast cancer metastasis is directly associated with breast cancer cell invasion. However, whether leptin could augment breast cancer cell invasion is not known. Here we showed that leptin increased the invasiveness and the matrix metallo-proteinase-2 (MMP-2) activity of the MCF-7 breast cancer cell line. Leptin stimulated the phosphorylation of extracellular signals regulated kinases, signal transducers and activators of transcription 3 and Jun N-terminal kinases (JNK); however, only inhibition of JNK decreased leptin-mediated activation of MMP-2. Furthermore, inhibition of JNK suppressed leptin-mediated breast cancer cell invasion. Here we report the novel findings that leptin increased invasion of breast cancer cells by activating JNK, resulting in increased MMP-2 activity.


Subject(s)
Breast Neoplasms/pathology , JNK Mitogen-Activated Protein Kinases/physiology , Leptin/physiology , Matrix Metalloproteinase 2/metabolism , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Humans , Leptin/pharmacology , MAP Kinase Signaling System/physiology , Neoplasm Invasiveness , Phosphorylation , Signal Transduction
5.
Breast Cancer Res Treat ; 114(2): 203-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18386173

ABSTRACT

High levels of the cyclooxygenase-2 (COX-2) protein have been associated with invasion and metastasis of breast tumors. Both prostaglandin E(2) (PGE(2)) and interleukin-8 (IL-8) have been shown to mediate the invasive activity of COX-2 in breast cancer cells. Here we expand these studies to determine how COX-2 uses PGE(2) and IL-8 to induce breast cancer cell invasion. We demonstrated that PGE(2) and IL-8 decreased the expression of the tumor suppressor protein Programmed Cell Death 4 (PDCD4). We hypothesized that suppression of PDCD4 expression is vital to the invasive activity of PGE(2) and IL-8. In MCF-7 cells overexpressing PDCD4 (MCF-7/PDCD4), PGE(2) and IL-8 failed to induce invasion, in contrast to the parental MCF-7 cells, thus indicating that PDCD4 blocks breast cancer cell invasion. MCF-7/PDCD4 cells produced higher levels of the Tissue Inhibitor of Metalloproteinases-2 (TIMP-2) than the parental cells. Silencing TIMP-2 mRNA in MCF-7/PDCD4 cells reversed the anti-invasive effects of PDCD4, allowing PGE(2) and IL-8 to induce the invasion of these cells. Here we report the novel findings that suppression of PDCD4 expression is vital for the invasive activity of COX-2 mediated by PGE(2) and IL-8, and that PDCD4 increases TIMP-2 expression to inhibit breast cancer cell invasion.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/pathology , RNA-Binding Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/genetics , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression/physiology , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Neoplasm Invasiveness , RNA Interference/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Cells, Cultured
6.
Breast Cancer Res ; 10(3): R44, 2008.
Article in English | MEDLINE | ID: mdl-18474097

ABSTRACT

INTRODUCTION: Tumor invasion and metastasis remain a major cause of mortality in breast cancer patients. High concentrations of nitric oxide (NO) suppress tumor invasion and metastasis in vivo. NO prodrugs generate large amounts of NO upon metabolism by appropriate intracellular enzymes, and therefore could have potential in the prevention and therapy of metastatic breast cancer. METHODS: The present study was designed to determine the effects of the NO-releasing prodrug O2-(2,4-dinitrophenyl) 1- [(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (JS-K) on breast cancer invasion and the mechanisms involved. MDA-MB-231, MDA-MB-231/F10, and MCF-7/COX-2 were the three breast cancer cell lines tested. NO levels were determined spectrophotometrically using a NO assay kit. Invasion and the expression of matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs were determined using Matrigel invasion assays, an MMP array kit and ELISAs. The activity and expression of extracellular signal-regulated kinase 1/2, p38, and c-Jun N-terminal kinase mitogen-activated protein kinases were determined using western blot analyses. RESULTS: Under conditions by which JS-K was not cytotoxic, JS-K significantly decreased (P < 0.05) the invasiveness of breast cancer cells across the Matrigel basement membrane, which was directly correlated with NO production. JS-43-126, a non-NO-releasing analog of JS-K, had no effect on NO levels or invasion. JS-K increased (P < 0.05) TIMP-2 production, and blocking TIMP-2 activity with a neutralizing antibody significantly increased (P < 0.05) the invasive activity of JS-K-treated cells across Matrigel. JS-K decreased p38 activity, whereas the activity and the expression of extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase were unaffected. CONCLUSION: We report the novel findings that JS-K inhibits breast cancer invasion across the Matrigel basement membrane, and NO production is vital for this activity. Upregulation of TIMP-2 production is one mechanism by which JS-K mediates its anti-invasive effects. JS-K and other NO prodrugs may represent an innovative biological approach in the prevention and treatment of metastatic breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Azo Compounds/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoplasm Invasiveness , Nitric Oxide/metabolism , Piperazines/pharmacology , Prodrugs/pharmacology , Tissue Inhibitor of Metalloproteinase-2/metabolism , Basement Membrane/metabolism , Cell Line, Tumor , Cell Proliferation , Collagen/chemistry , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Humans , Laminin/chemistry , Models, Chemical , Neoplasm Metastasis , Proteoglycans/chemistry
7.
J Exp Ther Oncol ; 7(4): 299-312, 2008.
Article in English | MEDLINE | ID: mdl-19227010

ABSTRACT

Inflammatory breast cancer (IBC) is the most aggressive form of locally advanced breast cancer (LABC) characterized by rapid growth and aggressive invasion with no selective therapies developed to treat IBC. Cyclooxygenase-2 (Cox-2), which produces prostaglandin E2 (PGE2) is known to be upregulated in primary IBC tumors and metastatic lesions, however the use of selective Cox-2 inhibitors has diminished due to cardiovascular side effects. One alternative approach to targeting Cox-2 enzyme activity is to block binding of the PGE2 ligand to its prostanoid (EP) receptors, which are designated as EP1, EP2, EP3, and EP4 and are members of a subfamily of G protein coupled receptors (GPCRs). While SUM149 IBC tumor cells and MCF-7 non-IBC breast tumor cells produce both EP2 and EP4 receptors, the invasive MDA-MB-231 non-IBC breast tumor cells produced low but detectable levels of these receptors. PGE2 and the EP4 agonist, PGE2 alcohol, stimulated significantly increased (p < 0.05) levels of proliferation and invasion by SUM149 IBC tumor cells, with no effect on proliferation of either of the two non-IBC breast tumor cell lines. In contrast, the EP2 agonist butaprost had no effect on proliferation or invasion of any cell line examined. The selective EP4 antagonist, GW627368X, induced inhibition of proliferation and invasion of human SUM149 IBC tumor cells beginning at 0.1 microM, with inhibition of proliferation and invasion by MDA-MB-231 non-IBC cells at higher concentrations of GW627368X. Molecular knockdown of the EP4 receptor was accomplished by stable transfection of an EP4 short hairpin RNA (shRNA) construct, with a clonally derived cell line designated as SUM149/Clone 1 exhibiting significantly slowed proliferation and diminished invasion compared to SUM149/Vector 5 which contained a scrambled shRNA control vector. This is the first report using both a selective pharmacologic inhibitor and a molecular shRNA knockdown approach to demonstrate that EP4 is directly involved in regulation of proliferation and invasion of IBC cells.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Receptors, Prostaglandin E/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Drug Screening Assays, Antitumor , Humans , Isoindoles/pharmacology , Models, Biological , Neoplasm Invasiveness , Receptors, Prostaglandin E, EP4 Subtype , Sulfonamides/pharmacology , Time Factors
8.
Int J Oncol ; 30(4): 785-92, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17332916

ABSTRACT

Cyclooxygenase-2 (COX-2) increases breast cancer cell invasion. Expression of various pro-angiogenic and pro-invasive factors has been correlated with high expression of COX-2. However, whether these factors are essential to COX-2-mediated breast cancer invasion, and the mechanisms by which COX-2 increases the expression of these factors are unknown. Our microarray results indicate that higher COX-2 expression was associated with increased levels of interleukin-8 (IL-8), a key factor in breast cancer invasion and metastasis. COX-2 overexpressing cells (MCF-7/COX-2), generated by transfecting COX-2-encoding plasmids into the poorly invasive MCF-7 breast cancer cells, were more invasive and produced higher IL-8 levels than the parental cells. To investigate the role of IL-8 in COX-2-mediated invasion, MCF-7 parental cells were incubated with IL-8. Exogenous IL-8 increased the invasiveness of MCF-7 cells. IL-8 is one pathway by which COX-2 mediates breast cancer invasion. Protein kinase A (PKA) and protein kinase C (PKC) are activated by COX-2 and are involved in IL-8 regulation. Inhibition of PKC, not PKA, decreased IL-8 production and invasion in MCF-7/COX-2 cells. Activation of PKC, not PKA, increased IL-8 production and invasion in MCF-7 cells. Thus, the invasive effects of COX-2 are mediated by PKC, not PKA. Activity of the urokinase-type plasminogen activator (uPA) was increased in MCF-7 cells by COX-2 overexpression or by the addition of a PKC activator or by IL-8. Inhibition of PKC decreased uPA activity in MCF-7/COX-2 cells. Furthermore, inhibition of uPA activity decreased the invasiveness of MCF-7/COX-2 cells, indicating that uPA was essential to COX-2-mediated invasion. Herein we demonstrate for the first time a detailed mechanism by which COX-2 increases breast cancer invasion: the PKC/IL-8/uPA pathway.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cyclooxygenase 2/metabolism , Interleukin-8/metabolism , Membrane Proteins/metabolism , Protein Kinase C/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Cyclooxygenase 2/genetics , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Neoplasm Invasiveness , Transfection
9.
Carcinogenesis ; 27(3): 568-77, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16199439

ABSTRACT

Breast cancer most frequently metastasizes to bone causing decreased quality of life and morbidity. Since current treatments are palliative, strategies to prevent bone metastases in breast cancer patients are required. There is substantial evidence indicating that high levels of nitric oxide (NO) suppress tumor growth and metastasis in vivo. We hypothesize that agents that produce high concentrations of NO could prevent the spread of breast cancer to bone. We previously demonstrated that the synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) produces high levels of NO via the induction of NO synthases. NO pro-drugs are designed to produce large amounts of NO without inducing NO synthases but upon metabolism by their intracellular targets. The objective of this study was to determine the effectiveness of 4-HPR and an NO pro-drug, diethylamineNONOate/AM (NONO-AM), in inhibiting the growth and invasiveness of bone metastatic breast cancer cells. Parental MDA-MB-231 breast cancer cells were resistant to 4-HPR-induced apoptosis at clinically relevant doses, whereas 4-HPR-induced apoptosis in a dose-dependent manner in MDA-MB-231/F10 bone metastatic breast cancer cells. Unlike 4-HPR, NONO-AM induced apoptosis in a dose-dependent manner in both parental MDA-MB-231 cells and F10 cells. The bone metastatic F10 cells were more sensitive to the anti-invasive effects of 4-HPR and NONO-AM than were MDA-MB-231 cells. Although suppression of matrix metalloprotease-9 activity may be one mechanism by which 4-HPR decreases the invasion of F10 cells, it does not appear to be the anti-invasion mechanism of NONO-AM. These in vitro results suggest that 4-HPR and NO pro-drugs may be effective chemopreventive agents against bone metastatic breast cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Fenretinide/pharmacology , Nitric Oxide/physiology , Dose-Response Relationship, Drug , Enzyme Induction , Female , Humans , Hydrazines/pharmacology , Neoplasm Invasiveness , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Nitrogen Oxides/pharmacology , Tumor Cells, Cultured
10.
Lab Invest ; 85(11): 1357-67, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16127422

ABSTRACT

Approximately 30-40% of estrogen receptor alpha (ERalpha)-positive breast tumors express high levels of the cyclooxygenase-2 (COX-2) protein, and these high levels have been associated with a poorer prognosis in breast cancer patients. We speculate that high levels of COX-2 induce drug resistance in ERalpha-positive breast tumors, thus reducing the survival rate of patients with such tumors. Human breast cancer cell lines that express high levels of COX-2 are generally ERalpha negative. To determine whether COX-2 induces drug resistance, plasmids encoding the COX-2 gene were stably transfected into ERalpha-positive MCF-7 human breast cancer cells (MCF-7/COX-2). MCF-7/COX-2 cells were resistant to the selective estrogen receptor modulator tamoxifen but not to its analog, raloxifene. MCF-7/COX-2 cells were also resistant to the retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) but not to its analog, all-trans retinoic acid. In contrast, the sensitivities of MCF-7/COX-2 cells to doxorubicin and paclitaxel were similar to those of the parental MCF-7 cells. We then determined which COX-2 product, prostaglandin E2 (PGE2) or prostaglandin F2alpha is involved in the COX-2-mediated drug resistance. PGE2, but not PGF2alpha, blocked the antiproliferative effects of tamoxifen and 4-HPR. Agonists that activate PGE2 receptors and their downstream kinase effectors, protein kinases A and C, also blocked the growth inhibitory effects of these drugs. Increased levels of Bcl-2 and Bcl-XL proteins have been reported in mammary tumors of COX-2 transgenic mice and in human colon cancer cell lines that have high levels of COX-2. However, we did not observe any changes in Bcl-2, Bcl-XL, or Bax expression induced by COX-2 or PGE2. Here we report the novel findings that COX-2 uses PGE2 to stimulate the activities of protein kinases A and C to induce selectively tamoxifen and 4-HPR resistance in ERalpha-positive breast cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Cyclooxygenase 2/metabolism , Fenretinide/antagonists & inhibitors , Membrane Proteins/metabolism , Selective Estrogen Receptor Modulators/metabolism , Tamoxifen/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Line, Tumor , Clone Cells , Drug Resistance, Neoplasm , Female , Humans , Inhibitory Concentration 50
11.
Carcinogenesis ; 26(5): 1000-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15695235

ABSTRACT

Women with germline mutations in the breast cancer susceptibility gene BRCA1 are at an increased risk of developing breast cancer. The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been shown to have a clinical chemopreventive activity in patients with premenopausal breast cancer. Since BRCA1 mutations are associated with an early-onset breast cancer, usually before menopause, we hypothesized that 4-HPR may be an effective chemopreventive agent against breast tumors exhibiting BRCA1 mutations. The objective of this study was to determine the effectiveness and mechanisms of action of 4-HPR and its phenylretinamide analogues in BRCA1-mutated breast cancer cells. At clinically relevant doses, 4-HPR induced apoptosis in human (HCC1937) and murine (W0069, W525) BRCA1-mutated breast cancer cells. Among the various phenylretinamides tested, N-(2-carboxyphenyl)retinamide (2-CPR) and 3-CPR significantly inhibited the growth of HCC1937 cells; however, they were not as potent as 4-HPR in this respect. We also determined the mechanisms by which 4-HPR induces apoptosis in BRCA1-mutated breast cancer cells. The extent to which 4-HPR induced apoptosis in BRCA1-mutated cells correlated with the increases in nitric oxide (NO) production and nitric oxide synthase (NOS) II and NOSIII expression. Use of a NOS inhibitor to block NO production suppressed the inhibitory effects of 4-HPR in all cell lines. These in vitro results suggest that 4-HPR may be an effective chemopreventive agent against breast tumors that exhibit BRCA1 mutations because of its ability to induce NO-mediated apoptosis in such tumors.


Subject(s)
Breast Neoplasms/drug therapy , Fenretinide/pharmacology , Genes, BRCA1 , Mutation , Apoptosis/drug effects , Breast Neoplasms/metabolism , Female , Fenretinide/analogs & derivatives , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Tumor Cells, Cultured
12.
Cancer Res ; 64(4): 1224-8, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14973114

ABSTRACT

We reported that HER2/neu reduces the sensitivity of breast cancer cells to N-(4-hydroxyphenyl)retinamide (4-HPR) by suppressing nitric oxide production. We show that HER2/neu uses Akt to induce cyclooxygenase-2 (COX-2) expression and that inhibition of Akt or COX-2 increases 4-HPR-induced apoptosis and nitric oxide production. Apoptosis induced by the 4-HPR and COX-2 inhibitor combination, although unaffected by an anti-HER2/neu antibody, was reversed by the COX-2 product prostaglandin E(2), indicating that COX-2 is a major mechanism by which HER2/neu suppresses 4-HPR apoptosis in breast cancer cells. Combining 4-HPR with COX-2 inhibitors may be a novel chemopreventive strategy against HER2/neu-overexpressing breast tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Fenretinide/pharmacology , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Protein Serine-Threonine Kinases , Receptor, ErbB-2/physiology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprost/pharmacology , Female , Humans , Membrane Proteins , Nitric Oxide/biosynthesis , Nitrobenzenes/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Sulfonamides/pharmacology
13.
Oncogene ; 22(43): 6739-47, 2003 Oct 02.
Article in English | MEDLINE | ID: mdl-14555987

ABSTRACT

The retinoid N-(4-hydroxyphenyl)retinamide (4-HPR also known as fenretinide) is a potent inducer of apoptosis in breast cancer cells. We observed a 4.5-fold reduction in 4-HPR-mediated apoptosis in MCF-7 breast cancer cells transfected with HER2/neu (MCF-7/HER2) as compared with the parental MCF-7 (MCF-7/WT) cells. Blocking HER2/neu with trastuzumab (Herceptin) led to a six-fold increase in 4-HPR-induced apoptosis in HER2/neu-overexpressing cells. These data indicate that HER2/neu reduces the sensitivity of breast cancer cells to 4-HPR. We showed previously that nitric oxide (NO) is essential for 4-HPR to induce apoptosis in breast cancer cells. The inhibitory effects of the 4-HPR and trastuzumab combination correlated with the amount of NO produced in HER2/neu-overexpressing cells. When a NO synthase (NOS) inhibitor was used to block NO production, decreased apoptosis by the 4-HPR and trastuzumab combination was observed. Furthermore, 4-HPR-mediated NOSII expression was lower in MCF-7/HER2 than MCF-7/WT cells, but was increased by trastuzumab in HER2/neu-overexpressing cells. Here we report the novel findings that HER2/neu reduces the ability of 4-HPR to induce apoptosis in breast cancer cells, and that one mechanism by which HER2/neu increases the resistance of breast cancer cells to 4-HPR is by decreasing NOSII-mediated NO production.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Fenretinide/metabolism , Nitric Oxide/metabolism , Receptor, ErbB-2/metabolism , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Blotting, Western , Cell Division , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Immunohistochemistry , Nitric Oxide Synthase/antagonists & inhibitors , Trastuzumab , Tretinoin/metabolism , Tumor Cells, Cultured
14.
Mol Cancer Ther ; 1(12): 1009-17, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12481423

ABSTRACT

N-(4-Hydroxyphenyl)retinamide (4-HPR) induces apoptosis in breast cancer cells; however, the molecular basis by which 4-HPR induces apoptosis is not well understood. In breast cancer cells, nitric oxide (NO) is predominantly an apoptotic inducer. Apoptotic agents, such as phorbol ester, tumor necrosis factor-alpha, and peptide hormones, have been shown to increase NO production in breast cancer cells. Therefore, we hypothesized that the production of No is vital for 4-HPR to induce apoptosis in breast cancer cells. We found that 4-HPR induced NO production in a dose-dependent manner in all of the breast cancer cell lines tested. The degree of growth inhibition and apoptotic induction by 4-HPR was directly correlated with the amount of NO produced. To prove that NO is essential for 4-HPR to induce apoptosis, breast cancer cells were coincubated with a competitive NO synthase (NOS) inhibitor, NG-monomethyl-L-arginine (L-NMMA), and 4-HPR, L-NMMA prevented 4-HPR from inducing inhibitory effects, indicating that NO is crucial for 4-HPR to induce its apoptotic effects in breast cancer cells. IFNs and tamoxifen (TAM) have been shown to potentiate 4-HPR effects in breast cancer cells. Both IFN-gamma and TAM enhanced the ability of 4-HPR to induce NO production in breast cancer cells, which was correlated with increased apoptosis. Alone, 4-HPR increased expression of both inducible NOS (NOSII) and endothelial NOS (NOSIII). When combined with 4-HPR, IFN-gamma and TAM enhanced NOSII expression. Thus, we have identified a novel mechanism by which 4-HPR induces apoptosis in breast cancer cells, i.e., by increasing NOS expression to induce NO production.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Fenretinide/pharmacology , Nitric Oxide/metabolism , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Nitric Oxide Synthase/metabolism , Tamoxifen/metabolism , Tamoxifen/pharmacology , Tumor Cells, Cultured , omega-N-Methylarginine/pharmacology
15.
Int J Cancer ; 101(3): 243-7, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12209974

ABSTRACT

4HPR, an analogue of ATRA, effectively induces growth inhibition and apoptosis in breast cancer cell lines and animal models but is ineffective against advanced human breast tumors. Different compounds, including tamoxifen, are currently being tested to increase 4HPR efficacy in the clinic. Here, we report that cyclosporin A selectively increases the ability of 4HPR, but not ATRA, to induce growth inhibition and apoptosis in ER(+) and ER(-) breast cancer cell lines. Increased apoptosis by the 4HPR and cyclosporin A combination was correlated with increased production of the free radical nitric oxide. Thus, the 4HPR and cyclosporin A combination may potentially be a novel therapeutic modality against breast tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cyclosporine/pharmacology , Fenretinide/pharmacology , Immunosuppressive Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Division/drug effects , Drug Synergism , Female , Humans , Nitric Oxide/metabolism , Receptors, Estrogen/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL