51
|
Nitric oxide: role in tumour biology and iNOS/NO-based anticancer therapies. Cancer Chemother Pharmacol 2011; 67:1211-24. [DOI: 10.1007/s00280-011-1654-4] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 04/14/2011] [Indexed: 01/10/2023]
|
52
|
McMurtry V, Saavedra JE, Nieves-Alicea R, Simeone AM, Keefer LK, Tari AM. JS-K, a nitric oxide-releasing prodrug, induces breast cancer cell death while sparing normal mammary epithelial cells. Int J Oncol 2011; 38:963-71. [PMID: 21271218 PMCID: PMC7295088 DOI: 10.3892/ijo.2011.925] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 11/19/2010] [Indexed: 11/06/2022] Open
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.
Collapse
Affiliation(s)
- Vanity McMurtry
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
53
|
HISHIKAWA K, NAKAGAWA H, MIYATA N. Nitric Oxide Donors Activated by Two-Photon Excitation. YAKUGAKU ZASSHI 2011; 131:317-24. [DOI: 10.1248/yakushi.131.317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Naoki MIYATA
- Graduate School of Pharmaceutical Science, Nagoya City University
| |
Collapse
|
54
|
Kostourou V, Cartwright JE, Johnstone AP, Boult JKR, Cullis ER, Whitley G, Robinson SP. The role of tumour-derived iNOS in tumour progression and angiogenesis. Br J Cancer 2010; 104:83-90. [PMID: 21139581 PMCID: PMC3039789 DOI: 10.1038/sj.bjc.6606034] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Background: Progressive tumour growth is dependent on the development of a functional tumour vasculature and highly regulated by growth factors and cytokines. Nitric oxide (NO) is a free radical, produced both by tumour and host cells, and functions as a signalling molecule downstream of several angiogenic factors. Both pro- and antitumourigenic properties have been attributed to NO. Methods: The expression of the inducible isoform of NO synthase (iNOS) was knocked down in the C6 glioma cell line using constitutive expression of antisense RNA, and the effect of tumour-derived NO on tumour progression and angiogenesis was investigated. Results: Tumours in which iNOS expression was decreased displayed significantly reduced growth rates compared with tumours derived from parental C6 cells. Quantitative non-invasive magnetic resonance imaging and fluorescence microscopy of tumour uptake of Hoechst 33342, and haematoxylin and eosin staining, revealed significantly impaired vascular development and function in antisense iNOS tumours compared with control in vivo, primarily associated with the more necrotic tumour core. Decreased iNOS expression had no effect on tumour VEGF expression. Conclusion: Nitric oxide derived from tumour iNOS is an important modulator of tumour progression and angiogenesis in C6 gliomas and further supports the therapeutic strategy of inhibiting iNOS for the treatment of cancer.
Collapse
Affiliation(s)
- V Kostourou
- Division of Basic Medical Sciences, St George's University of London, Cranmer Terrace, London SW17 ORE, UK
| | | | | | | | | | | | | |
Collapse
|
55
|
Nitric oxide-mediated tumoricidal activity of murine microglial cells. Transl Oncol 2010; 3:380-8. [PMID: 21151477 DOI: 10.1593/tlo.10208] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 08/16/2010] [Accepted: 08/18/2010] [Indexed: 02/03/2023] Open
Abstract
Experimental metastases in the brain of mice are infiltrated by microglia, and parabiosis experiments of green fluorescent protein (GFP(+)) and GFP(-) mice revealed that these microglia are derived from circulating monocytes (GFP(+), F4/80(+), and CD68(+)). These findings raised the question as to whether microglia (specialized macrophages) possess tumoricidal activity. C8-B4 murine microglia cells were incubated in vitro in medium (control) or in medium containing both lipopolysaccharide and interferon-γ. Control microglia were not tumoricidal against a number of murine and human tumor cells, whereas lipopolysaccharide/interferon-γ-activated microglia lysed murine and human tumor cells by release of nitric oxide. Parallel experiments with murine peritoneal macrophages produced identical results. Neither activated microglia nor activated macrophages lysed nontumorigenic murine or human cells. Collectively, these data demonstrate that brain metastasis-associated microglia are derived from circulating mononuclear cells and exhibit selective and specific tumoricidal activity.
Collapse
|
56
|
Muntané J, la Mata MD. Nitric oxide and cancer. World J Hepatol 2010; 2:337-44. [PMID: 21161018 PMCID: PMC2999298 DOI: 10.4254/wjh.v2.i9.337] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 09/02/2010] [Accepted: 09/09/2010] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) is a lipophilic, highly diffusible and short-lived physiological messenger which regulates a variety of important physiological responses including vasodilation, respiration, cell migration, immune response and apoptosis. NO is synthesized by three differentially gene-encoded NO synthase (NOS) in mammals: neuronal NOS (nNOS or NOS-1), inducible NOS (iNOS or NOS-2) and endothelial NOS (eNOS or NOS-3). All isoforms of NOS catalyze the reaction of L-arginine, NADPH and oxygen to NO, L-citrulline and NADP. NO may exert its cellular action by cGMP-dependent as well as by cGMP-independent pathways including postranslational modifications in cysteine (S-nitrosylation or S-nitrosation) and tyrosine (nitration) residues, mixed disulfide formation (S-nitrosoglutathione or GSNO) or promoting further oxidation protein stages which have been related to altered protein function and gene transcription, genotoxic lesions, alteration of cell-cycle check points, apoptosis and DNA repair. NO sensitizes tumor cells to chemotherapeutic compounds. The expression of NOS-2 and NOS-3 has been found to be increased in a variety of human cancers. The multiple actions of NO in the tumor environment is related to heterogeneous cell responses with particular attention in the regulation of the stress response mediated by the hypoxia inducible factor-1 and p53 generally leading to growth arrest, apoptosis or adaptation.
Collapse
Affiliation(s)
- Jordi Muntané
- Jordi Muntané, Liver Research Unit, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), "Reina Sofia" University Hospital, Cordoba E-14004, Spain
| | | |
Collapse
|
57
|
Chin MP, Deen WM. Prediction of nitric oxide concentrations in melanomas. Nitric Oxide 2010; 23:319-26. [PMID: 20854923 DOI: 10.1016/j.niox.2010.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 06/16/2010] [Accepted: 09/13/2010] [Indexed: 11/30/2022]
Abstract
The presence of iNOS and nitrotyrosine in cutaneous melanomas has been correlated with poor survival rates of patients, suggesting that NO plays a role in the tumor pathophysiology. However, the concentrations of NO that melanoma cells are exposed to in vivo have been unknown. To provide cell kinetic data for use in predicting those concentrations, synthesis and consumption of NO was examined in A375 melanoma cells. Nitric oxide synthesis was undetectable. The rate of intracellular NO consumption was determined by continuous monitoring of NO concentrations following injection of NO solutions in a closed chamber. After correcting for autoxidation and consumption from media-generated O(2)(-), the rate constant obtained for cellular consumption was 7.1±1.1 s(-1). This information was combined with previous data on macrophage NO kinetics to develop a mathematical model to predict NO levels in cutaneous melanomas. Synthesis of NO by macrophages in the stroma was found to give a maximum concentration at the tumor periphery of 0.2 μM. Because of the high rates of cellular consumption, the elevation in NO concentration is predicted to be very localized, approximately 90% of the concentration decay occurring within 30 μm of the tumor edge. High NO concentrations at the periphery of a melanoma may contribute to metastasis by stimulating cell proliferation, inhibiting apoptosis, or acting as a lymphangiogenic factor.
Collapse
Affiliation(s)
- Melanie P Chin
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
58
|
Sikora AG, Gelbard A, Davies MA, Sano D, Ekmekcioglu S, Kwon J, Hailemichael Y, Jayaraman P, Myers JN, Grimm EA, Overwijk WW. Targeted inhibition of inducible nitric oxide synthase inhibits growth of human melanoma in vivo and synergizes with chemotherapy. Clin Cancer Res 2010; 16:1834-44. [PMID: 20215556 PMCID: PMC2858983 DOI: 10.1158/1078-0432.ccr-09-3123] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE Aberrant expression of inflammatory molecules, such as inducible nitric oxide (NO) synthase (iNOS), has been linked to cancer, suggesting that their inhibition is a rational therapeutic approach. Whereas iNOS expression in melanoma and other cancers is associated with poor clinical prognosis, in vitro and in vivo studies suggest that iNOS and NO can have both protumor and antitumor effects. We tested the hypothesis that targeted iNOS inhibition would interfere with human melanoma growth and survival in vivo in a preclinical model. EXPERIMENTAL DESIGN We used an immunodeficient non-obese diabetic/severe combined immunodeficient xenograft model to test the susceptibility of two different human melanoma lines to the orally-given iNOS-selective small molecule antagonist N(6)-(1-iminoethyl)-l-lysine-dihydrochloride (L-nil) with and without cytotoxic cisplatin chemotherapy. RESULTS L-nil significantly inhibited melanoma growth and extended the survival of tumor-bearing mice. L-nil treatment decreased the density of CD31+ microvessels and increased the number of apoptotic cells in tumor xenografts. Proteomic analysis of melanoma xenografts with reverse-phase protein array identified alterations in the expression of multiple cell signaling and survival genes after L-nil treatment. The canonical antiapoptotic protein Bcl-2 was downregulated in vivo and in vitro after L-nil treatment, which was associated with increased susceptibility to cisplatin-mediated tumor death. Consistent with this observation, combination therapy with L-nil plus cisplatin in vivo was more effective than either drug alone, without increased toxicity. CONCLUSIONS These data support the hypothesis that iNOS and iNOS-derived NO support tumor growth in vivo and provide convincing preclinical validation of targeted iNOS inhibition as therapy for solid tumors.
Collapse
Affiliation(s)
- Andrew G. Sikora
- UT MD Anderson Cancer Center: Dept. of Melanoma Medical Oncology, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
- UT MD Anderson Cancer Center: Dept. of Head and Neck Surgery, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
- Departments of Otolaryngology, Immunobiology, Oncological Sciences, and Dermatology, Mount Sinai School of Medicine, One Gustave L. Levy. Place, Box 1189, New York, NY 10029
| | - Alexander Gelbard
- UT MD Anderson Cancer Center: Dept. of Melanoma Medical Oncology, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
- UT MD Anderson Cancer Center: Dept. of Head and Neck Surgery, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
- Bobby Alford Department of Otolaryngology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
| | - Michael A. Davies
- UT MD Anderson Cancer Center: Dept. of Melanoma Medical Oncology, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
- UT MD Anderson Cancer Center: Dept. Of Systems Biology, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - Daisuke Sano
- UT MD Anderson Cancer Center: Dept. of Head and Neck Surgery, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - Suhendan Ekmekcioglu
- UT MD Anderson Cancer Center: Dept. of Experimental Therapeutics, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - John Kwon
- UT MD Anderson Cancer Center: Dept. of Experimental Therapeutics, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - Yared Hailemichael
- CORRESPONDING AUTHOR: WWO (; Phone: 713-563-5294; Fax: 713-563-3424).. AGS (; Phone: 212-659-9516; Fax: 212-369-5701)
| | - Padmini Jayaraman
- Departments of Otolaryngology, Immunobiology, Oncological Sciences, and Dermatology, Mount Sinai School of Medicine, One Gustave L. Levy. Place, Box 1189, New York, NY 10029
| | - Jeffrey N. Myers
- UT MD Anderson Cancer Center: Dept. of Head and Neck Surgery, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - Elizabeth A. Grimm
- UT MD Anderson Cancer Center: Dept. of Experimental Therapeutics, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| | - Willem W. Overwijk
- UT MD Anderson Cancer Center: Dept. of Melanoma Medical Oncology, 1515 Holcombe Blvd, Unit 430, Houston, TX 77030
| |
Collapse
|
59
|
Prabhu V, Guruvayoorappan C. Nitric oxide: pros and cons in tumor progression. Immunopharmacol Immunotoxicol 2010; 32:387-92. [DOI: 10.3109/08923970903440192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
60
|
Teruya T, Tatemoto H, Konishi T, Tako M. Structural characteristics and in vitro macrophage activation of acetyl fucoidan from Cladosiphon okamuranus. Glycoconj J 2009; 26:1019-28. [PMID: 19219547 DOI: 10.1007/s10719-008-9221-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 11/06/2008] [Accepted: 12/09/2008] [Indexed: 11/24/2022]
Abstract
We investigated a structural characteristics of acetyl fucoidan (CAF) isolated from commercially cultured Cladosiphon okamuranus. The CAF-induced macrophage activation and its signaling pathways in murine macrophage cell line, RAW 264.7 were also investigated. From the results of methylation analysis, CAF consisted of alpha-1-->3 linked L: -fucosyl residues and substituted sulfate and acetyl groups at C-4 on the main chain. CAF induced production of nitric oxide (NO), tumor necrosis factor-alpha and interleukin-6 in RAW 264.7 cells. Sulfate and acetyl groups of CAF involved in CAF-induced NO production. Neutralizing anti-Toll-like receptor 4 (TLR4), anti-CD14 and anti-scavenger receptor class A (SRA) but not anti-complement receptor type 3 monoclonal antibodies decreased CAF-induced NO production. The results of immunoblot analysis indicated that CAF activated mitogen-activated protein kinases (MAPKs) such as p38 MAPK, extracellular signal-regulated kinase (ERK)1/2 and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK). SB203580 (p38 MAPK inhibitor) and SP600125 (SAPK/JNK inhibitor), but not U0126 (MAPK/ERK kinase 1/2 inhibitor) decreased CAF-induced NO production. The results suggested that CAF induced macrophage activation through membrane receptors TLR4, CD14 and SRA, and MAPK signaling pathways.
Collapse
Affiliation(s)
- Takeshi Teruya
- The United Graduate School of Agricultural Sciences, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-0065, Japan.
| | | | | | | |
Collapse
|
61
|
Palmieri G, Capone M, Ascierto ML, Gentilcore G, Stroncek DF, Casula M, Sini MC, Palla M, Mozzillo N, Ascierto PA. Main roads to melanoma. J Transl Med 2009; 7:86. [PMID: 19828018 PMCID: PMC2770476 DOI: 10.1186/1479-5876-7-86] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 10/14/2009] [Indexed: 12/12/2022] Open
Abstract
The characterization of the molecular mechanisms involved in development and progression of melanoma could be helpful to identify the molecular profiles underlying aggressiveness, clinical behavior, and response to therapy as well as to better classify the subsets of melanoma patients with different prognosis and/or clinical outcome. Actually, some aspects regarding the main molecular changes responsible for the onset as well as the progression of melanoma toward a more aggressive phenotype have been described. Genes and molecules which control either cell proliferation, apoptosis, or cell senescence have been implicated. Here we provided an overview of the main molecular changes underlying the pathogenesis of melanoma. All evidence clearly indicates the existence of a complex molecular machinery that provides checks and balances in normal melanocytes. Progression from normal melanocytes to malignant metastatic cells in melanoma patients is the result of a combination of down- or up-regulation of various effectors acting on different molecular pathways.
Collapse
Affiliation(s)
- Giuseppe Palmieri
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche (CNR), Sassari, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Manuele MG, Arcos MLB, Davicino R, Ferraro G, Cremaschi G, Anesini C. Mechanism of the antiproliferative action of limonene on a lymphoma cell line: participation of nitric oxide. antiproliferative action of limonene on a lymphoma cell line. Phytother Res 2009; 23:1011-7. [DOI: 10.1002/ptr.2743] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
63
|
Avci A, Tüzüner-Oncül AM, Gökcan MK, Namuslu M, Oztürk A, Durak I. Nitric oxide metabolism in cancerous and non-cancerous oral gingivomucosal tissues: possible implications of nitric oxide in cancer process. J Oral Pathol Med 2009; 38:304-6. [PMID: 19141063 DOI: 10.1111/j.1600-0714.2008.00719.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Nitric oxide (NO) is a molecule that plays various roles in the body tissues. NO plays important roles in vasodilatation, platelet aggregation, cytokine stimulation, neurotransmission, immune function, etc. NO also exerts dual functions as an oxidant and antioxidant substance depending on its concentrations and environmental conditions. In this study, we aimed to examine possible correlation between NO levels and NO synthase (NOS) activity in the patients with oral cancer. MATERIALS AND METHODS The study included 19 tissues from human subjects (11 malign and eight benign lesions). RESULTS NO level and NOS activity were found decreased in the malign lesions compared with those of the benign ones. CONCLUSIONS In conclusion, two suggestions can be made; first, decreased NO synthesis may be an attempt to suppress angiogenesis, which is known to provide more essential nutrients to malign lesions and/or second, malign lesions may suppress NO production to be capable of creating more rapid proliferation as it has been known that NO is also a powerful free radical inducing apoptosis.
Collapse
Affiliation(s)
- A Avci
- Department of Biochemistry, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
64
|
Reeves KJ, Reed MW, Brown NJ. Is nitric oxide important in photodynamic therapy? JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2009; 95:141-7. [DOI: 10.1016/j.jphotobiol.2009.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 02/05/2009] [Accepted: 02/10/2009] [Indexed: 10/21/2022]
|
65
|
Heller A. Apoptosis-inducing high (.)NO concentrations are not sustained either in nascent or in developed cancers. ChemMedChem 2009; 3:1493-9. [PMID: 18759245 DOI: 10.1002/cmdc.200800257] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nitric oxide ((.)NO) induces apoptosis at high concentrations by S-nitrosating proteins such as glyceraldehyde-3-phosphate dehydrogenase. This literature analysis revealed that failure to sustain high (.)NO concentrations is common to all cancers. In cervical, gastric, colorectal, breast, and lung cancer, the cause of this failure is the inadequate expression of inducible nitric oxide synthase (iNOS), resulting from the inhibition of iNOS expression by TGF-beta1 at the mRNA level. In bladder, renal, and prostate cancer, the reason for the insufficient (.)NO levels is the depletion of arginine, resulting from arginase overexpression. Arginase competes with iNOS for arginine, catalyzing its hydrolysis to ornithine and urea. In gliomas and ovarian sarcomas, low (.)NO levels are caused by inhibition of iNOS by N-chlorotaurine, produced by infiltrating neutrophils. Stimulated neutrophils express myeloperoxidase, catalyzing H2O2 oxidation of Cl- to HOCl, which N-chlorinates taurine at its concentration of 19 mM in neutrophils. In squamous cell carcinomas of the skin, ovarian cancers, lymphomas, Hodgkin's disease, and breast cancers, low (.)NO concentrations arise from the inhibition of iNOS by N-bromotaurine, produced by eosinophil-peroxidase-expressing infiltrating eosinophils. Eosinophil peroxidase catalyzes the H2O2 oxidation of Br- to HOBr, which N-brominates taurine to N-bromotaurine at its concentration of 15 mM in eosinophils. In microvascularized tumors, the (.)NO concentration is further depleted; (.)NO is rapidly consumed by red blood cells (RBCs) through S-nitrosation of RBC glutathione and hemoglobin, and by oxidation to nitrate by RBC oxyhemoglobin. Angiogenesis-inhibiting antibodies are currently used to treat cancers; their mode of action is not, as previously thought, reduction of the tumor O2 or nutrient supply. They actually decrease the loss of (.)NO to RBCs.
Collapse
Affiliation(s)
- Adam Heller
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
66
|
Yang Z, Yang S, Misner BJ, Chiu R, Liu F, Meyskens FL. Nitric oxide initiates progression of human melanoma via a feedback loop mediated by apurinic/apyrimidinic endonuclease-1/redox factor-1, which is inhibited by resveratrol. Mol Cancer Ther 2009; 7:3751-60. [PMID: 19074850 DOI: 10.1158/1535-7163.mct-08-0562] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is well recognized that nitric oxide (NO) is involved in tumor progression, including melanoma. Measurement of proliferative and metastatic capacity by MTS and Matrigel invasion assays, respectively, was done and showed that NO-treated melanoma cells exhibited a higher capacity compared with control, especially metastatic Lu1205 cells. Apurinic/apyrimidinic endonuclease-1/redox factor-1 (APE/Ref-1) is a multifunctional protein and its role in tumor biology has attracted considerable attention. To determine whether APE/Ref-1 plays a role in mediating NO stimulation of melanoma progression, we investigated the effect of DETA/NO on levels of APE/Ref-1 and related downstream targets [activator protein-1 (AP-1)/JunD, matrix metalloproteinase-1 (MMP-1), Bcl-2, and inducible nitric oxide synthase (iNOS)] by Western blot and reverse transcription-PCR analysis. Following DETA/NO treatment, APE/Ref-1 and other downstream molecules were induced. Knockdown of APE/Ref-1 or AP-1/JunD by specific small interfering RNA markedly reversed the induction by NO stress of target proteins. These results present evidence for the existence of a functional feedback loop contributing to progression and metastasis of melanoma cells. Resveratrol has been shown to be an APE/Ref-1 inhibitor and significant decreases in AP-1/JunD, MMP-1, Bcl-2, and iNOS protein levels occurred after exposure to resveratrol. This phenolic antioxidant may be an appropriate choice for combining with other compounds that develop resistance by up-regulation of these molecules.
Collapse
Affiliation(s)
- Zhen Yang
- Chao Family Comprehensive Cancer Center, University of California-Irvine School of Medicine, Orange, California, USA
| | | | | | | | | | | |
Collapse
|
67
|
Yang GY, Taboada S, Liao J. Induced nitric oxide synthase as a major player in the oncogenic transformation of inflamed tissue. Methods Mol Biol 2009; 512:119-156. [PMID: 19347276 DOI: 10.1007/978-1-60327-530-9_8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nitric oxide (NO) is a free radical that is involved in the inflammatory process and carcinogenesis. There are four nitric oxide synthase enzymes involved in NO production: induced nitric oxide synthase (iNOS), endothelial NO synthase (eNOS), neural NO synthase (nNOS), and mitochondrial NOS. iNOS is an inducible and key enzyme in the inflamed tissue. Recent literatures indicate that NO as well as iNOS and eNOS can modulate cancer-related events including nitro-oxidative stress, apoptosis, cell cycle, angio-genesis, invasion, and metastasis. This chapter focuses on linking NO/iNOS/eNOS to inflammation and carcinogenesis from experimental evidence to potential targets on cancer prevention and treatment.
Collapse
Affiliation(s)
- Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | |
Collapse
|
68
|
McCarthy HO, Coulter JA, Robson T, Hirst DG. Gene therapy via inducible nitric oxide synthase: a tool for the treatment of a diverse range of pathological conditions. J Pharm Pharmacol 2008; 60:999-1017. [PMID: 18644193 DOI: 10.1211/jpp.60.8.0007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide (NO(.)) is a reactive nitrogen radical produced by the NO synthase (NOS) enzymes; it affects a plethora of downstream physiological and pathological processes. The past two decades have seen an explosion in the understanding of the role of NO(.) biology, highlighting various protective and damaging modes of action. Much of the controversy surrounding the role of NO(.) relates to the differing concentrations generated by the three isoforms of NOS. Both calcium-dependent isoforms of the enzyme (endothelial and neuronal NOS) generate low-nanomolar/picomolar concentrations of NO(.). By contrast, the calcium-independent isoform (inducible NOS (iNOS)) generates high concentrations of NO(.), 2-3 orders of magnitude greater. This review summarizes the current literature in relation to iNOS gene therapy for the therapeutic benefit of various pathological conditions, including various states of vascular disease, wound healing, erectile dysfunction, renal dysfunction and oncology. The available data provide convincing evidence that manipulation of endogenous NO(.) using iNOS gene therapy can provide the basis for future clinical trials.
Collapse
Affiliation(s)
- Helen O McCarthy
- School of Pharmacy, McClay Research Centre, Queen's University, Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | | | | | | |
Collapse
|
69
|
Jensen-Jarolim E, Achatz G, Turner MC, Karagiannis S, Legrand F, Capron M, Penichet ML, Rodríguez JA, Siccardi AG, Vangelista L, Riemer AB, Gould H. AllergoOncology: the role of IgE-mediated allergy in cancer. Allergy 2008; 63:1255-66. [PMID: 18671772 PMCID: PMC2999743 DOI: 10.1111/j.1398-9995.2008.01768.x] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Epidemiological studies have suggested inverse associations between allergic diseases and malignancies. As a proof of concept for the capability of immunoglobulin E (IgE) to destruct tumor cells, several experimental strategies have evolved to specifically target this antibody class towards relevant tumor antigens. It could be demonstrated that IgE antibodies specific to overexpressed tumor antigens have been superior to any other immunoglobulin class with respect to antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) reactions. In an alternative approach, IgE nonspecifically attached to tumor cells proved to be a powerful adjuvant establishing tumor-specific immune memory. Active Th2 immunity could also be achieved by applying an oral immunization regimen using mimotopes, i.e. epitope mimics of tumor antigens. The induced IgE antibodies could be cross-linked by live tumor cells leading to tumoricidic mediator release. Thus, IgE antibodies may not only act in natural tumor surveillance, but could possibly also be exploited for tumor control in active and passive immunotherapy settings. Thereby, eosinophils, mast cells and macrophages can be armed with the cytophilic IgE and become potent anti-tumor effectors, able to trace viable tumor cells in the tissues. It is strongly suggested that the evolving new field AllergoOncology will give new insights into the role of IgE-mediated allergy in malignancies, possibly opening new avenues for tumor therapy.
Collapse
Affiliation(s)
- E Jensen-Jarolim
- Department of Pathophysiology, Center of Physiology, Pathophysiology and Immunology, Medical University Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Abdelmagid SA, Too CKL. Prolactin and estrogen up-regulate carboxypeptidase-d to promote nitric oxide production and survival of mcf-7 breast cancer cells. Endocrinology 2008; 149:4821-8. [PMID: 18535109 DOI: 10.1210/en.2008-0145] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Carboxypeptidase-D (CPD) and carboxypeptidase-M (CPM) release C-terminal arginine (Arg) from polypeptides, and both are present in the plasma membrane. Cell-surface CPD increases intracellular Arg, which is converted to nitric oxide (NO). We have reported that prolactin (PRL) regulated CPD mRNA levels in MCF-7 breast cancer cells. This study examined PRL/17beta-estradiol (E2) regulation of CPD/CPM expression, and the role of CPD in NO production for survival of MCF-7 cells. We showed that PRL or E2 up-regulated CPD mRNA and protein expression. PRL/E2 increased CPD mRNA levels by 3- to 5-fold but had no effect on CPM. In Arg-free DMEM, exogenous L-Arg or substrate furylacryloyl-Ala-Arg (Fa-Ala-Arg) increased NO levels and cell survival, measured using 4,5-diaminofluorescein diacetate and the MTS assay, respectively. In the presence of Fa-Ala-Arg, NO production was enhanced by PRL and/or E2 but inhibited by CPD/CPM-specific inhibitor, 2-mercaptomethyl-3-guanidinoethylthio-propanoic acid (MGTA). MGTA also decreased MCF-7 cell survival. In Arg-free medium, annexin-V staining showed that apoptotic MCF-7 cells (approximately 60%) were rescued by Fa-Ala-Arg (25%) or diethylamine/NO (10%). Finally, CPD or CPM gene expression was knocked down with small interfering (si) CPD or siCPM, respectively, with nontargeting siNT as controls. In Arg-free DMEM, the stimulatory effect of Fa-Ala-Arg on NO production was inhibited by siCPD only, showing that CPD depletion inhibited Fa-Ala-Arg cleavage. Furthermore, more than 60% of siCPD-transfectants were apoptotic, and L-Arg, not Fa-Ala-Arg, significantly decreased apoptosis to 32% (P<or=0.05). Thus, CPD gene knockdown did not affect L-Arg uptake, which protected cells from apoptosis. In summary, PRL/E2-induced cell-surface CPD released Arg from extracellular substrates, increased intracellular NO, promoted survival and inhibited apoptosis of MCF-7 cells.
Collapse
Affiliation(s)
- Salma A Abdelmagid
- Department of Biochemistry and Molecular Biology, Sir Charles Tupper Medical Building, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada B3H 1X5
| | | |
Collapse
|
71
|
Wink DA, Ridnour LA, Hussain SP, Harris CC. The reemergence of nitric oxide and cancer. Nitric Oxide 2008; 19:65-7. [PMID: 18638716 PMCID: PMC2565861 DOI: 10.1016/j.niox.2008.05.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 05/16/2008] [Indexed: 01/19/2023]
Affiliation(s)
- David A Wink
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
72
|
Cytotoxicity of goniothalamin enantiomers in renal cancer cells: involvement of nitric oxide, apoptosis and autophagy. Chem Biol Interact 2008; 176:143-50. [PMID: 18771661 DOI: 10.1016/j.cbi.2008.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Revised: 07/30/2008] [Accepted: 08/04/2008] [Indexed: 02/03/2023]
Abstract
Goniothalamin is a styryllactone synthesized by plants of the genus Goniothalamus. The biological activities of this molecule, particularly its anti-protozoan, anti-fungal, and larvicidal properties, have received considerable attention. In this work, we investigated the action of the natural and synthetic enantiomers (R)-goniothalamin (1) and (S)-goniothalamin (ent-1) on cell viability, nitric oxide synthase (NOS) expression and activity, and the expression of selected proteins involved in apoptosis and autophagy in renal cancer cells. Both compounds were cytotoxic and decreased the mitochondrial function of renal cancer cells. However, the enantiomers differentially affected the expression/activity profiles of some signaling pathway mediators. Ent-1 (4 nM) was more potent than 1 (6.4 microM) in inhibiting constitutive NOS activity (54% and 59% inhibition, respectively), and both enantiomers decreased the protein expression of neuronal and endothelial NOS, as assessed by western blotting. Ent-1 and 1 caused down-regulation of Ras and TNFR1 and inhibition of protein serine/threonine phosphatase 2A (PP2A). Compound 1 markedly down-regulated Bcl2, an anti-apoptotic protein, and also induced PARP cleavage. Despite inducing an expressive down-regulation of Bax, ent-1 was also able to induce PARP cleavage. These results suggest that these compounds caused apoptosis in renal cancer cells. Interestingly, ent-1 enhanced the expression of LC3, a typical marker of autophagy. NFkappaB was down-regulated in 1-treated cells. Overall, these results indicate that the anti-proliferative activity of the two enantiomers on renal cancer cells involved distinct signaling pathways, apoptosis and autophagy as dominant responses towards 1 and ent-1, respectively.
Collapse
|
73
|
Cyclin A expression is associated with apoptosis and mitosis in murine 3-methylcholanthrene-induced fibrosarcomas. ACTA ACUST UNITED AC 2008; 61:41-9. [PMID: 18621517 DOI: 10.1016/j.etp.2008.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 05/26/2008] [Accepted: 05/26/2008] [Indexed: 11/23/2022]
Abstract
The chemical carcinogen MCA induces fibrosarcoma and tissue damage at the injection site. Despite the importance of ROS in the development of cancer, little is known about the pattern of expression of ROS in MCA-induced fibrosarcomas. To gain some insight into the biological significance of iNOS and Cu/Zn-SOD, comparative immunohistochemical analyses were performed to characterize their expression in MCA-induced fibrosarcomas. Cyclin A is overexpressed in various tumors, but its expression in MCA-induced fibrosarcoma in mice and its correlation to mitosis and apoptosis are unclear. The presence of apoptotic cell death was evaluated using the TUNEL method and findings were compared with cyclin A expression and mitotic count of fibrosarcomas. Subcutaneous application of MCA caused fibrosarcoma development in 14 of 20 mice (70%) in 26 weeks. Limited cytoplasmic Cu/Zn-SOD and iNOS immunostainings were detected in 13 of 14 and 9 of 14 tumors with median immunoreactive scores of 2 and 1, respectively. Prominent nuclear cyclin A immunostaining and TUNEL-positive reactions were seen in all the fibrosarcoma cases. Cyclin A immunoreaction significantly correlated with the TUNEL index (P<0.01) and MC (P<0.001). The present findings show a low level of iNOS expression in neoplastic cells indicating limited synthesizing capacity of tumor cells. Limited Cu/Zn-SOD reaction could be associated with an imbalance in between pro-oxidant/antioxidant levels. Furthermore, it was shown that cyclin A is overexpressed in MCA-induced fibrosarcomas and possibly plays a significant role in the pathogenesis of fibrosarcomas. Cyclin A could be useful for detecting the S phase of the cell cycle and could also indicate that cyclin A may induce S phase arrest associated with apoptosis in the MCA-induced fibrosarcomas.
Collapse
|
74
|
Cheng A, Wan F, Jin Z, Wang J, Xu X. Nitrite oxide and inducible nitric oxide synthase were regulated by polysaccharides isolated from Glycyrrhiza uralensis Fisch. JOURNAL OF ETHNOPHARMACOLOGY 2008; 118:59-64. [PMID: 18434050 DOI: 10.1016/j.jep.2008.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 01/09/2008] [Accepted: 03/06/2008] [Indexed: 05/26/2023]
Abstract
Water-soluble polysaccharide(Glycyrrhiza polysaccharide, GP) was isolated from Glycyrrhiza uralensis Fisch, with glycosidic units were composed of alpha (1-4) linked D-glucana. We demonstrated that GP significantly induces nitric oxides (NO) production and inducible NO synthase (iNOS) transcription in peritoneal macrophages. Moreover, iNOS mRNA expression was strongly induced by GP. NO in the culture supernatant was measured by Griess reaction, the production of iNOS was determined by commercially available iNOS kit. GP (10-400 microg/ml) alone increased significantly NO and iNOS production in macrophages. Macrophages simultaneously treated with GP plus lipopolysaccharide (LPS)/interferon-gamma (IFN-gamma) increased NO and iNOS production as compared to that of GP treatments alone. The production of NO and iNOS in macrophages pretreated with LPS followed by GP was higher than that of treatment with GP and LPS simultaneously. Using RT-PCR reveals that GP may provide a second triggering signal for the expression of iNOS mRNA. Thus, the iNOS-mediated NO synthesis in response to GP may be one of the mechanisms whereby this herbal medicine elicits its therapeutic effects.
Collapse
Affiliation(s)
- Anwei Cheng
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | | | | | | | | |
Collapse
|
75
|
Menon C, Bauer TW, Kelley ST, Raz DJ, Bleier JI, Patel K, Steele K, Prabakaran I, Shifrin A, Buerk DG, Sehgal CM, Fraker DL. Tumoricidal activity of high-dose tumor necrosis factor-alpha is mediated by macrophage-derived nitric oxide burst and permanent blood flow shutdown. Int J Cancer 2008; 123:464-475. [PMID: 18449880 DOI: 10.1002/ijc.23499] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This study investigates the role of tumor nitric oxide (NO) and vascular regulation in tumor ulceration following high-dose tumor necrosis factor-alpha (TNF) treatment. Using TNF-responsive (MethA) and nonresponsive (LL2) mouse tumors, tumor NO concentration was measured with an electrochemical sensor and tumor blood flow by Doppler ultrasound. Mice were also pretreated with a selective inducible nitric oxide synthase (iNOS) inhibitor, 1400 W. Tumors harvested from TNF-treated mice were cryosectioned and immunostained for murine macrophages, or/and iNOS. MethA tumor-bearing mice were depleted of macrophages. Pre- and post-TNF tumor NO levels were measured continuously, and mice were followed for gross tumor response. In MethA tumors, TNF caused a 96% response rate, and tumor NO concentration doubled. Tumor blood flow decreased to 3% of baseline by 4 hr and was sustained at 24 hr and 10 days post-TNF. Selective NO inhibition with 1400 W blocked NO rise and decreased response rate to 38%. MethA tumors showed tumor infiltration by macrophages post-TNF and the pattern of macrophage immunostaining overlapped with iNOS immunostaining. Depletion of macrophages inhibited tumor NO increase and response to TNF. LL2 tumors had a 0% response rate to TNF and exhibited no change in NO concentration. Blood flow decreased to 2% of baseline by 4 hr, recovered to 56% by 24 hr and increased to 232% by 10 days. LL2 tumors showed no infiltration by macrophages post-TNF. We conclude that TNF causes tumor infiltrating, macrophage-derived iNOS-mediated tumor NO rise and sustained tumor blood flow shutdown, resulting in tumor ulceration in the responsive tumor.
Collapse
Affiliation(s)
| | - Todd W Bauer
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Scott T Kelley
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Dan J Raz
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Joshua I Bleier
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Krina Patel
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Kirsten Steele
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | | | | | - Donald G Buerk
- Department of Physiology, University of Pennsylvania, Philadelphia, PA
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Douglas L Fraker
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
76
|
Stefano GB, Kream RM, Mantione KJ, Sheehan M, Cadet P, Zhu W, Bilfinger TV, Esch T. Endogenous morphine/nitric oxide-coupled regulation of cellular physiology and gene expression: implications for cancer biology. Semin Cancer Biol 2008; 18:199-210. [PMID: 18203618 PMCID: PMC2432462 DOI: 10.1016/j.semcancer.2007.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Accepted: 12/05/2007] [Indexed: 12/19/2022]
Abstract
Cancer is a simplistic, yet complicated, process that promotes uncontrolled growth. In this regard, this unconstrained proliferation may represent primitive phenomena whereby cellular regulation is suspended or compromised. Given the new empirical evidence for a morphinergic presence and its profound modulatory actions on several cellular processes it is not an overstatement to hypothesize that morphine may represent a key chemical messenger in the process of modulating proliferation of diverse cells. This has been recently demonstrated by the finding of a novel opiate-alkaloid selective receptor subtype in human multilineage progenitor cells (MLPC). Adding to the significance of morphinergic signaling are the findings of its presence in plant, invertebrate and vertebrate cells, which also have been shown to synthesize this messenger as well. Interestingly, we and others have shown that some cancerous tissues contain morphine. Furthermore, in medullary histolytic reticulosis, which is exemplified by cells having hyperactivity, the mu3 (mu3) opiate select receptor was not present. Thus, it would appear that morphinergic signaling has inserted itself in many processes taking a long time to evolve, including those regulating the proliferation of cells across diverse phyla.
Collapse
Affiliation(s)
- George B Stefano
- Neuroscience Research Institute, State University of New York - SUNY College at Old Westbury, P.O. Box 210, Old Westbury, NY 11568, USA.
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Simeone AM, McMurtry V, Nieves-Alicea R, Saavedra JE, Keefer LK, Johnson MM, Tari AM. TIMP-2 mediates the anti-invasive effects of the nitric oxide-releasing prodrug JS-K in breast cancer cells. Breast Cancer Res 2008; 10:R44. [PMID: 18474097 PMCID: PMC2481491 DOI: 10.1186/bcr2095] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Revised: 04/07/2008] [Accepted: 05/12/2008] [Indexed: 11/22/2022] Open
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.
Collapse
Affiliation(s)
- Ann-Marie Simeone
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vanity McMurtry
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - René Nieves-Alicea
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joseph E Saavedra
- Basic Research Program, SAIC-Frederick, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Larry K Keefer
- Laboratory of Comparative Carcinogenesis, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Marcella M Johnson
- Department of Biostatistics & Applied Mathematics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ana M Tari
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
78
|
Leon L, Jeannin JF, Bettaieb A. Post-translational modifications induced by nitric oxide (NO): implication in cancer cells apoptosis. Nitric Oxide 2008; 19:77-83. [PMID: 18474258 DOI: 10.1016/j.niox.2008.04.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 04/16/2008] [Accepted: 04/16/2008] [Indexed: 11/28/2022]
Abstract
Post-translational modifications of proteins can regulate the balance between survival and cell death signals. It is increasingly recognized that nitric oxide (NO) and reactive oxygen species (ROS)-induced post-translational modifications could play a role in cell death. This review provides an introduction of current knowledge of NO proteins modifications promoting or inhibiting cell death with special attention in cancer cells.
Collapse
Affiliation(s)
- Lissbeth Leon
- EPHE, Laboratoire d'immunologie et immunothérapie des cancers, Inserm U866, Dijon, F-21000, France.
| | | | | |
Collapse
|
79
|
NCX 4040, an NO-donating acetylsalicylic acid derivative: efficacy and mechanisms of action in cancer cells. Nitric Oxide 2008; 19:225-36. [PMID: 18472019 DOI: 10.1016/j.niox.2008.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 04/14/2008] [Accepted: 04/14/2008] [Indexed: 12/12/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) have repeatedly shown to be effective in tumor prevention, but important side-effects limit their wide clinical use. Nitric oxide-releasing derivatives (NO-NSAIDs) are a promising class of compounds synthesized by combining a classic NSAID molecule with an NO-releasing moiety to counteract side-effects. These new chemical entities exhibit a significantly higher activity and much lower toxicity with respect to the parental drug. In the present paper, we report the results obtained from in in vitro experimental systems aimed to evaluate the activity and mechanisms of action of the novel NO-releasing aspirin derivative, NCX 4040. The in vitro studies were carried out on a panel of human colon (LoVo, LoVo Dx, WiDr, LRWZ), bladder (HT1376, MCR), and pancreatic (Capan-2, MIA PaCa-2, T3M4) cancer cell lines. With regard to colon cancer, NCX 4040 activity was also investigated in vitro in combination with drugs currently used in clinical practice and was validated in vivo on tumor-bearing mice xenografted with the aforementioned colon cancer cell lines. The in vitro studies showed a high cytotoxic activity of NCX 4040 in all tumor histotypes and demonstrated the pivotal role of the NO component in drug activity. It was also observed that NCX 4040 exerts a pro-apoptotic activity via a mitochondria-dependent pathway. Moreover, the in vivo studies on xenografted mice further confirmed the antitumor efficacy and low toxicity of NCX 4040 in colon cancer and highlighted its role as sensitizing agent of oxaliplatin cytotoxicity.
Collapse
|
80
|
Morbidelli L, Donnini S, Ziche M. Nitric Oxide in Tumor Angiogenesis. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
81
|
Yamaguchi H, Noshita T, Kidachi Y, Umetsu H, Hayashi M, Komiyama K, Funayama S, Ryoyama K. Isolation of Ursolic Acid from Apple Peels and Its Specific Efficacy as a Potent Antitumor Agent. ACTA ACUST UNITED AC 2008. [DOI: 10.1248/jhs.54.654] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Toshiro Noshita
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| | - Yumi Kidachi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| | - Hironori Umetsu
- Laboratory of Food Chemistry, Department of Life Sciences, Junior College, Gifu Shotoku Gakuen University
| | | | | | - Shinji Funayama
- Department of Kampo Pharmaceutical Sciences, Nihon Pharmaceutical University
| | - Kazuo Ryoyama
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| |
Collapse
|
82
|
Turek M, Padilla M, Argyle DJ. Evaluation of the gene for inducible nitric oxide synthase as a radiosensitizer under hypoxic and oxic conditions. Vet Comp Oncol 2007; 5:250-5. [DOI: 10.1111/j.1476-5829.2007.00138.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
83
|
Korean mistletoe lectin (KML-IIU) and its subchains induce nitric oxide (NO) production in murine macrophage cells. J Biomed Sci 2007; 15:197-204. [DOI: 10.1007/s11373-007-9210-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 09/12/2007] [Indexed: 11/25/2022] Open
|
84
|
Yamaguchi H, Kidachi Y, Umetsu H, Ryoyama K. Ras/myc-transformed serum-free mouse embryo cells under simulated inflammatory and infectious conditions increase levels of nitric oxide and matrix metalloproteinase-9 without a direct association between them. Mol Cell Biochem 2007; 306:43-51. [PMID: 17660954 DOI: 10.1007/s11010-007-9552-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2007] [Accepted: 07/12/2007] [Indexed: 10/23/2022]
Abstract
Inflammatory and infectious conditions were simulated in cultures of ras/myc-transformed serum-free mouse embryo (ras/myc SFME) cells, using interferon-gamma (IFN-gamma, 100 units/ml) and lipopolysaccharide (LPS, 0.5 microg/ml) co-treatment for 24 h, to investigate their effects on the expression of inducible nitric oxide synthase (iNOS) mRNA and the production of NO. Aminoguanidine (AG, 1 mM; an NOS inhibitor) along with IFN-gamma and LPS, S-nitroso-N-acetyl-DL-penicillamine (SNAP, 100 microM; an NO donor) and/or (+/-)-N-[(E)-4-Ethyl-2-[(Z)-hydroxyimino]-5-nitro-3-hexene-1-yl]-3-pyridine carboxamide (NOR4, 100 microM; an NO donor), were also added to analyze the possible association of NO with matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1). Co-treatment of cells with IFN-gamma and LPS increased iNOS mRNA expression, NO production, MMP-9 mRNA expression, and 105 kDa MMP-9 production. Additional treatment with the NOS inhibitor AG inhibited NO production, but did not down-regulate the expression of MMP-9 mRNA or 105 kDa MMP-9. The NO donors SNAP and NOR4 did not affect the expression of MMP-9 mRNA, 105 kDa MMP-9 or TIMP-1 mRNA. These results suggest that ras/myc SFME cells respond to infectious and inflammatory conditions and can enhance malignancy as cancer cells due to their increased levels of NO and MMP-9 production, but that NO is not directly associated with MMP-9 in these cells.
Collapse
Affiliation(s)
- Hideaki Yamaguchi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University, 2-3-1 Kobata, Aomori 030-0943, Japan.
| | | | | | | |
Collapse
|
85
|
Zhang XF, Cui Y, Huang JJ, Zhang YZ, Nie Z, Wang LF, Yan BZ, Tang YL, Liu Y. Immuno-stimulating properties of diosgenyl saponins isolated from Paris polyphylla. Bioorg Med Chem Lett 2007; 17:2408-13. [PMID: 17350838 DOI: 10.1016/j.bmcl.2007.02.039] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2007] [Revised: 02/12/2007] [Accepted: 02/15/2007] [Indexed: 10/23/2022]
Abstract
The effects of three diosgenyl saponins isolated from Paris polyphylla on the immuno-stimulating activity in relation to phagocytosis, respiratory burst, and nitric oxide production in mouse macrophage cells RAW 264.7 have been investigated. Our results showed that all three diosgenyl saponins significantly enhanced phagocytic activity that increased with the concentration of saponins to reach a maximum, and then tended to decrease with higher concentrations. Saponins with sugar moiety directly induced respiratory burst response in RAW 264.7 cells that increased with the concentrations and reached a maximum, then decreased with higher concentrations after 2-h incubations, however, diosgenin had no PMA-triggered respiratory burst response. Treatment of RAW 264.7 cells with saponins with sugar moiety for 24-h caused a significant increase in the production of nitric oxide, while diosgenin had no effect at all. Consequently, relationship between molecular structures of three diosgenyl saponins and their immunomodulatory activities was discussed, and a possible mechanism of immuno-stimulating function of diosgenyl saponins was accordingly explored.
Collapse
Affiliation(s)
- Xiu-feng Zhang
- State Key laboratory for SCUSS, Institute of Chemistry, The Chinese Academy of Sciences, Beijing 100080, China
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Zou F, Liu Y, Liu L, Wu K, Wei W, Zhu Y, Wu J. Retinoic acid activates human inducible nitric oxide synthase gene through binding of RARalpha/RXRalpha heterodimer to a novel retinoic acid response element in the promoter. Biochem Biophys Res Commun 2007; 355:494-500. [PMID: 17306764 DOI: 10.1016/j.bbrc.2007.01.178] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Accepted: 01/30/2007] [Indexed: 02/03/2023]
Abstract
Human inducible nitric oxide synthase (hiNOS) catalyzes nitric oxide (NO) which has a significant effect on tumor suppression and cancer therapy. Here we revealed the detailed molecular mechanism involved in the regulation of hiNOS expression induced by retinoic acid (RA). We showed that RARalpha/RXRalpha heterodimer was important in hiNOS promoter activation, hiNOS protein expression, and NO production. Serial deletion and site-directed mutation analysis revealed two half-sites of retinoic acid response element (RARE) spaced by 5bp located at -172 to -156 in the hiNOS promoter. EMSA and ChIP assays demonstrated that RARalpha/RXRalpha directly bound to this RARE of hiNOS promoter. Our results suggested the identification of a novel RARE in the hiNOS promoter and the roles of the nuclear receptors (RARalpha/RXRalpha) in the induction of hiNOS by RA.
Collapse
Affiliation(s)
- Fang Zou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, PR China
| | | | | | | | | | | | | |
Collapse
|
87
|
Kang KW, Wagley Y, Kim HW, Pokharel YR, Chung YY, Chang IY, Kim JJ, Moon JS, Kim YK, Nah SY, Kang HS, Oh JW. Novel role of IL-6/SIL-6R signaling in the expression of inducible nitric oxide synthase (iNOS) in murine B16, metastatic melanoma clone F10.9, cells. Free Radic Biol Med 2007; 42:215-27. [PMID: 17189827 DOI: 10.1016/j.freeradbiomed.2006.10.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Revised: 09/15/2006] [Accepted: 10/08/2006] [Indexed: 12/27/2022]
Abstract
Inducible nitric oxide synthase (iNOS) has been shown to be frequently expressed in melanomas; up-regulation of this enzyme is though to be associated with tumor progression. In this study, we investigated whether diverse cytokines such as: IL-6, TNF-alpha, IL-1beta, IFN-gamma and IL6RIL6 (a highly active fusion protein of the soluble form of the IL-6R (sIL-6R) and IL-6) enhance the iNOS gene expression in B16/F10.9 murine metastatic melanoma cells. An increase at iNOS expression and NO production was observed with the co-treatment of IL6RIL6 plus TNF-alpha. Gel shift and reporter gene analyses revealed that IL6RIL6 selectively activated AP-1; while TNF-alpha increased the activities of both NF-kappaB and AP-1. Persistent activation of AP-1 was also seen in cells treated with IL6RIL6 plus TNF-alpha. Stimulation of cells with IL6RIL6/TNF-alpha resulted in the activation of mitogen-activated protein kinases (MAPK) such as c-Jun N-terminal kinase (JNK) and p38, and the abrogation by pretreatment with JNK or p38 MAPK inhibitor. IL6RIL6 or IL6RIL6/TNFalpha-inducible AP-1 binding increase was supershifted by anti-c-Jun or c-Fos antibodies, and the activation of c-Jun and c-Fos was dependent on JNK and p38, respectively. These results suggest that IL-6/sIL-6R/gp130 complex signaling has an unexpected positive effect on iNOS gene expression through JNK/p38 MAPK mediated-AP-1 activation in melanoma cells.
Collapse
Affiliation(s)
- Keon Wook Kang
- College of Pharmacy, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Yao JC, Zhang JX, Rashid A, Yeung SCJ, Szklaruk J, Hess K, Xie K, Ellis L, Abbruzzese JL, Ajani JA. Clinical and In vitro Studies of Imatinib in Advanced Carcinoid Tumors. Clin Cancer Res 2007; 13:234-40. [PMID: 17200360 DOI: 10.1158/1078-0432.ccr-06-1618] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Effective systemic therapy options for carcinoid tumors are lacking. We conducted in vitro studies and a phase II clinical trial to explore the activity of imatinib in carcinoid tumors. EXPERIMENTAL DESIGN Cells of the human bronchial carcinoid cell line NCI-H727 and the human pancreatic carcinoid cell line BON-1 were treated with increasing concentrations of imatinib using standard procedures to assess in vitro growth-inhibitory activity. A clinical trial using a two-stage phase II design to assess the response rate and safety profile of imatinib at a dose of 400 mg given twice daily in patients with advanced carcinoid tumors was completed. RESULTS In both cell lines, there was a dose- and time-dependent cytotoxic effect. The clinical trial enrolled 27 evaluable patients. Median duration on trial was 16 weeks. One patient had a partial response, 17 had stable disease, and 9 had progressive disease by the Response Evaluation Criteria in Solid Tumors criteria. Median progression-free survival time was 24 weeks. Median overall survival is 36 months. Seven patients who achieved a biochemical response had a superior progression-free survival time compared with patients without biochemical response (115 weeks compared with 24 weeks; P = 0.003). An increase in plasma basic fibroblast growth factor was associated with a shorter progression-free survival duration (P = 0.02). CONCLUSIONS Our data suggest that imatinib is active in vitro and has a modest clinical activity in carcinoid patients. Changes in tumor markers may help select patients who are likely to benefit from therapy.
Collapse
Affiliation(s)
- James C Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Mocellin S, Bronte V, Nitti D. Nitric oxide, a double edged sword in cancer biology: Searching for therapeutic opportunities. Med Res Rev 2007; 27:317-52. [PMID: 16991100 DOI: 10.1002/med.20092] [Citation(s) in RCA: 337] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) is a pleiotropic molecule critical to a number of physiological and pathological processes. The last decade has witnessed major advances in dissecting NO biology and its role in cancer pathogenesis. However, the complexity of the interactions between different levels of NO and several aspects of tumor development/progression has led to apparently conflicting findings. Furthermore, both anti-NO and NO-based anticancer strategies appear effective in several preclinical models. This paradoxical dichotomy is leaving investigators with a double challenge: to determine the net impact of NO on cancer behavior and to define the therapeutic role of NO-centered anticancer strategies. Only a comprehensive and dynamic view of the cascade of molecular and cellular events underlying tumor biology and affected by NO will allow investigators to exploit the potential antitumor properties of drugs interfering with NO metabolism. Available data suggest that NO should be considered neither a universal target nor a magic bullet, but rather a signal transducer to be modulated according to the molecular makeup of each individual cancer and the interplay with conventional antineoplastic agents.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Oncological and Surgical Sciences, School of Medicine, University of Padova, Padova, Italy.
| | | | | |
Collapse
|
90
|
Hollenbaugh JA, Dutton RW. IFN-γ Regulates Donor CD8 T Cell Expansion, Migration, and Leads to Apoptosis of Cells of a Solid Tumor. THE JOURNAL OF IMMUNOLOGY 2006; 177:3004-11. [PMID: 16920936 DOI: 10.4049/jimmunol.177.5.3004] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously reported that IFN-gamma secreted by donor cytotoxic T cell 1 (Tc1) cells was the most important factor in promoting EG7 (an OVA transfection the EL4 thymoma) rejection in mice. In this study, we show that the ability of the host to respond to Tc1-secreted IFN-gamma is critical for promoting acute tumor rejection, while host production of IFN-gamma is not important. CFSE-labeled wild-type and IFN-gamma-deficient Tc1 cells divide rapidly in secondary lymphoid organs, indicating no defect in rate of cell division. However, wild-type Tc1 cells accumulate to significantly greater numbers in the tumor than deficient Tc1 cells. Hosts injected with wild-type Tc1 effectors had more T cells within the tumor at day 4, had higher levels of MCP-1, IFN-gamma-inducible protein-10, MIP-1alpha, and MIP-1beta mRNA transcripts, had greater numbers of CD11b+ and Gr-1+ cells within the tumor, and had massive regions of tumor cell apoptosis as compared with IFN-gamma knockout Tc1 cell-treated hosts. NO has a cytostatic effect on EG7 growth in vitro, and NO is important for tumor eradication by day 22. These observations are compatible with a model in which the donor CD8 Tc1 effectors expand rapidly in the host, migrate to the tumor site, and induce the secretion of a number of chemokines that in turn recruit host cells that then attack the tumor.
Collapse
|
91
|
Thomas DD, Ridnour LA, Espey MG, Donzelli S, Ambs S, Hussain SP, Harris CC, DeGraff W, Roberts DD, Mitchell JB, Wink DA. Superoxide fluxes limit nitric oxide-induced signaling. J Biol Chem 2006; 281:25984-93. [PMID: 16829532 DOI: 10.1074/jbc.m602242200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Independently, superoxide (O2-) and nitric oxide (NO) are biologically important signaling molecules. When co-generated, these radicals react rapidly to form powerful oxidizing and nitrating intermediates. Although this reaction was once thought to be solely cytotoxic, herein we demonstrate using MCF7, macrophage, and endothelial cells that when nanomolar levels of NO and O2- were produced concomitantly, the effective NO concentration was established by the relative fluxes of these two radicals. Differential regulation of sGC, pERK, HIF-1alpha, and p53 were used as biological dosimeters for NO concentration. Introduction of intracellular- or extracellular-generated O2- during NO generation resulted in a concomitant increase in oxidative intermediates with a decrease in steady-state NO concentrations and a proportional reduction in the levels of sGC, ERK, HIF-1alpha, and p53 regulation. NO responses were restored by addition of SOD. The intermediates formed from the reactions of NO with O2- were non-toxic, did not form 3-nitrotyrosine, nor did they elicit any signal transduction responses. H2O2 in bolus or generated from the dismutation of O2- by SOD, was cytotoxic at high concentrations and activated p53 independent of NO. This effect was completely inhibited by catalase, suppressed by NO, and exacerbated by intracellular catalase inhibition. We conclude that the reaction of O2- with NO is an important regulatory mechanism, which modulates signaling pathways by limiting steady-state levels of NO and preventing H2O2 formation from O2-.
Collapse
Affiliation(s)
- Douglas D Thomas
- Tumor Biology Section, Radiation Biology Branch, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
Nitric oxide (NO) and nitric oxide synthases are ubiquitous in malignant tumours and are known to exert both pro- and anti-tumour effects. We summarize our current understanding of the role of NO in tumour progression, especially in relation to angiogenesis and vascular functions. We also discuss potential strategies for cancer treatment that modulate NO production and/or its downstream signalling pathways.
Collapse
Affiliation(s)
- Dai Fukumura
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.
| | | | | |
Collapse
|
93
|
Ridnour LA, Thomas DD, Donzelli S, Espey MG, Roberts DD, Wink DA, Isenberg JS. The biphasic nature of nitric oxide responses in tumor biology. Antioxid Redox Signal 2006; 8:1329-37. [PMID: 16910780 DOI: 10.1089/ars.2006.8.1329] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The dual or biphasic responses of cancer to nitric oxide (NO) arise from its concentration dependent ability to regulate tumor growth, migration, invasion, survival, angiogenesis, and metastasis. The outcome of these various NO-dependent processes is dictated by several factors including NO flux, the chemical redox environment, and the duration of NO exposure. Further, it was recently discovered that an NO-induced redox flux in vascular endothelial cells hypersensitizes these cells to the antiangiogenic effects of thrombospondin-1. This suggests a novel treatment paradigm for targeting tumor-driven angiogenesis that combines redox modulation with mimetic derivatives of thrombospondin-1. This article discusses the biphasic nature of NO in cancer biology and the implications of NO-driven redox flux for modulation of tumor-stimulated angiogenesis, growth, and metastasis.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Tumor Biology Section, Radiation Biology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
94
|
Abstract
Unlike other types of cancer, tumors of the breast are greatly influenced by steroid hormones. The effect of estrogen and progesterone depends on the presence of their specific receptors and these constitute important parameters in determining the aggressiveness of the tumor, the feasibility of certain therapies and the prediction of relapse. The molecular mechanisms of steroid hormone action have not been fully elucidated but recent findings implicate the nitric oxide (NO) pathway in some of these effects. Both hormones can regulate the nitric oxide synthases (NOS) and, in turn, the NO produced has profound consequences on tumor cell homeostasis. On one hand, estrogen increases the activity of endothelial NOS (eNOS or NOSIII), while progesterone activates inducible NOS (iNOS or NOSII) expression. The data presented suggest that the low levels of NO produced by NOSIII mediate the proliferative effect of estrogen. On the other hand, the increase in apoptosis in response to progesterone could implicate the high levels of NO produced by induction of NOSII expression. Understanding of the mechanisms and interactions of steroid hormones with the NO pathway could lead to the development of new approaches and strategies for the effective treatment of breast cancer.
Collapse
Affiliation(s)
- Alena Pance
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
95
|
Tao J, Tu YT, Li JW, Feng AP, Huang CZ, Wu Y, Shen GX. Endogenous production of nitric oxide contributes to proliferation effect of vascular endothelial growth factor-induced malignant melanoma cell. Clin Exp Dermatol 2006; 31:94-9. [PMID: 16309495 DOI: 10.1111/j.1365-2230.2005.01922.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The objectives of this study were to observe the effect of overexpression of vascular endothelial growth factor (VEGF) on the proliferation of the malignant melanoma (MM) cell line A375, and to study the role of nitric oxide (NO) in this process and the mechanism of VEGF induced-A375 cell proliferation. The VEGF(165) cDNA was transfected into A375 cells by electroporation. VEGF mRNA and protein in A375 cells were detected by RT-PCR and ELISA. The proliferation of A375 cells was assessed by cell counting and MTT assay. Protein expression of iNOS, eNOS and nNOS was detected by Western blotting. NO production in A375 cell supernatant was measured by the nitrate reductase method. VEGF mRNA in A375 cells was significantly increased 72 h and 96 h after transfection of VEGF(165) cDNA, as were VEGF protein, NO and iNOS levels. However, protein expression of eNOS and nNOS was not detected in either transfected or untransfected cells. Proliferation of A375 cells transfected with VEGF(165) cDNA was enhanced. The nitric oxide synthase inhibitor l-NAME could dose-dependently inhibit the proliferation of A375 cells evoked by VEGF. These results indicate that VEGF enhances the expression of iNOS in A375 cells and results in an increase in NO formation, which may be important in the process of VEGF-induced proliferation of A375 cells.
Collapse
Affiliation(s)
- J Tao
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | | | | | | | | | | | | |
Collapse
|
96
|
Shen SC, Lin CW, Lee HM, Chien LL, Chen YC. Lipopolysaccharide plus 12-o-tetradecanoylphorbol 13-acetate induction of migration and invasion of glioma cells in vitro and in vivo: Differential inhibitory effects of flavonoids. Neuroscience 2006; 140:477-89. [PMID: 16580779 DOI: 10.1016/j.neuroscience.2006.02.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 01/10/2006] [Accepted: 02/11/2006] [Indexed: 11/19/2022]
Abstract
In an earlier study, we reported that nitric oxide is involved in lipopolysaccharide plus 12-o-tetradecanoylphorbol 13-acetate-induced malignant transformation via increases in metalloproteinase 9 enzyme activity and inducible nitric oxide synthase gene expression in rat glioma C6 cells, however the mechanism has remained undefined. Lipopolysaccharide plus 12-o-tetradecanoylphorbol 13-acetate, but not lipopolysaccharide or 12-o-tetradecanoylphorbol 13-acetate alone, induced transformation in glioma C6 cells (but not in human glioblastoma cells GBM-8401 cells) without affecting their viability. An increase in inducible nitric oxide synthase protein expression, nitric oxide production, and metalloproteinase 9 enzyme activity is identified lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate-treated C6 cells, however lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate and 12-o-tetradecanoylphorbol 13-acetate (but not lipopolysaccharide) addition shows the similar inductive pattern on metalloproteinase 9 enzyme activity without affecting inducible nitric oxide synthase protein expression and nitric oxide production in GBM-8401 cells. Treatment of C6 cells with lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate increases the expression of phosphorylated extracellular regulated protein kinases and Jun N-terminal kinases, but not p38, proteins, and an addition of the extracellular regulated protein kinases inhibitor PD98059 or Jun N-terminal kinases inhibitors SP600125, but not the p38 inhibitor SB203580, significantly blocked lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate-induced inducible nitric oxide synthase protein expression and metalloproteinase 9 enzyme activity accompanied by blocking morphological transformation in C6 cells. Among 19 structurally related flavonoids, kaempferol and wogonin exhibit significant inhibitory effects on lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate-induced morphological transformation and colony formation, and attenuation of inducible nitric oxide synthase, phosphorylated extracellular regulated protein kinases protein expression, and metalloproteinase 9 enzyme activity was observed. 2'-OH flavone at a dose of 100 microM inhibition of lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate-induced events via apoptosis induction is identified. Furthermore, lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate, but not lipopolysaccharide or 12-o-tetradecanoylphorbol 13-acetate, induces tumoral invasion and migration in vitro and in vivo, and those are blocked by kaempferol and wogonin addition. These data suggest that combination of lipopolysaccharide and 12-o-tetradecanoylphorbol 13-acetate promotes tumoral progression via activating metalloproteinase 9 enzyme activity and inducible nitric oxide synthase gene expression, which is located downstream of mitogen-activated protein kinases activation, in rat glioma cells C6. Kaempferol and wogonin exhibit effective inhibitory effects on lipopolysaccharide/12-o-tetradecanoylphorbol 13-acetate-induced events, and thus possess the potential for further development.
Collapse
Affiliation(s)
- S C Shen
- Department of Dermatology, School of Medicine, Taipei Medical University, and Department of Dermatology, Taipei Municipal Wan-Fang Hospital-Affiliated to Taipei Medical University, 111 Xinglong Road, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
97
|
Uğar-Cankal D, Ozmeric N. A multifaceted molecule, nitric oxide in oral and periodontal diseases. Clin Chim Acta 2006; 366:90-100. [PMID: 16387291 DOI: 10.1016/j.cca.2005.10.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2005] [Revised: 10/19/2005] [Accepted: 10/20/2005] [Indexed: 11/28/2022]
Abstract
Nitric oxide (NO) is a molecule with multiple effects on different tissues. NO takes important roles in vasodilatation, bacterial challenge and cytokine stimulation, regulation of mineralized tissue function, neurotransmission, and platelet aggregation, etc. However, under pathological conditions, NO has damaging effects. NO is synthesized by NO synthases (NOS) and inducible isoform of NOS (iNOS) is closely related to the pathophysiological characteristics of inflammatory diseases such as periodontal diseases. The expression of iNOS has been investigated in salivary gland-related diseases, temporomandibular joint disorders and oral cancer as well. The beneficial and damaging effects of NO in diseases related with periodontal, dental and maxillofacial area are discussed in this review. The biological pathways involved with NO and NO inhibitors may be good drug targets to have a role in the future management of patients with diseases in orofacial region.
Collapse
Affiliation(s)
- Dilek Uğar-Cankal
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Gazi University, Biskek caddesi 84.sokak 06510 Emek, Ankara, Turkey.
| | | |
Collapse
|
98
|
Okada F, Tazawa H, Kobayashi T, Kobayashi M, Hosokawa M. Involvement of reactive nitrogen oxides for acquisition of metastatic properties of benign tumors in a model of inflammation-based tumor progression. Nitric Oxide 2006; 14:122-9. [PMID: 16125421 DOI: 10.1016/j.niox.2005.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2005] [Revised: 06/16/2005] [Accepted: 06/20/2005] [Indexed: 11/30/2022]
Abstract
The cells of a weakly tumorigenic and non-metastatic murine fibrosarcoma (QR-32) are converted into highly malignant tumors (acquiring metastatic potential) once they have grown in vivo after being co-implanted with gelatin sponge which induces inflammation. In the present study, we examined whether nitric oxide (NO) is involved in the inflammation-based tumor progression by administrating a specific inhibitor to inducible nitric oxide synthase, aminoguanidine (AG). First, we co-implanted 1 x 10(5) QR-32 cells with gelatin sponge (10 x 5 x 3 mm piece) into a subcutaneous space in C57BL6 mice. Administration of AG in drinking water (1%) had started 2 days before the tumor implantation and continued until the termination of the experiment. The incidence of tumor formation and the tumor growth did not differ between AG-treated group and -untreated group. On day 28, we excised the arising tumors to establish culture cell lines for evaluation of their acquisition of metastatic phenotype in other normal mice. Metastasis incidence and the number of metastatic colonies were significantly reduced in the tumor cell lines obtained from AG-treated mice compared to those from non-treated mice (p < 0.05). Immunohistochemical analysis demonstrated that inducible nitric oxide synthase and nitrotyrosine in the inflamed lesion were reduced in the AG-administered mice. However, intensity of 8-hydroxy-2-deoxyguanosine was not different between the groups. These results showed that nitric oxide and its reactive nitrogen oxide species cooperatively play a pivotal role in the progression of benign tumor cells in inflamed lesions.
Collapse
Affiliation(s)
- Futoshi Okada
- Department of Biomolecular Function, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan.
| | | | | | | | | |
Collapse
|
99
|
Lee KY, Lee MH, Chang IY, Yoon SP, Lim DY, Jeon YJ. Macrophage activation by polysaccharide fraction isolated from Salicornia herbacea. JOURNAL OF ETHNOPHARMACOLOGY 2006; 103:372-8. [PMID: 16183225 DOI: 10.1016/j.jep.2005.08.037] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2005] [Revised: 07/12/2005] [Accepted: 08/16/2005] [Indexed: 05/04/2023]
Abstract
We demonstrate that polysaccharides isolated from Salicornia herbacea (Salicornia polysaccharides, SPS) significantly induces nitric oxide (NO) production and inducible NO synthase (iNOS) transcription through the activation of nuclear factor-kappaB/Rel (NF-kappaB/Rel). SPS dose-dependently induced the production of NO in isolated mouse peritoneal macrophages and RAW 264.7, a mouse macrophage-like cell line. Moreover, iNOS gene expression was strongly induced by SPS in RAW 264.7 cells. To further investigate the mechanism responsible for the induction of iNOS gene expression, we investigated the effect of SPS on the activation of transcription factors including NF-kappaB/Rel and Oct, whose binding sites were located in the promoter of iNOS gene. Treatment of RAW 264.7 cells with SPS produced strong induction of NF-kappaB/Rel-dependent reporter gene expression, whereas Oct-dependent gene expression was not affected by SPS. Nuclear translocation and DNA binding activity of NF-kappaB/Rel was significantly induced by SPS. The treatment with NF-kappaB SN50, an inhibitor of NF-kappaB/Rel nuclear translocation, effectively inhibited the activation of NF-kappaB/Rel binding complexes and NO production. In conclusion, we demonstrate that SPS stimulates macrophages to express iNOS gene through the activation of NF-kappaB/Rel.
Collapse
Affiliation(s)
- Kun Yeong Lee
- Department of Pharmacology, College of Medicine, Chosun University, Kwangju, Republic of Korea
| | | | | | | | | | | |
Collapse
|
100
|
Olson MV, Lee J, Zhang F, Wang A, Dong Z. Inducible nitric oxide synthase activity is essential for inhibition of prostatic tumor growth by interferon-beta gene therapy. Cancer Gene Ther 2006; 13:676-85. [PMID: 16470211 DOI: 10.1038/sj.cgt.7700941] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have previously reported that adenoviral vector-mediated interferon (IFN)-beta gene therapy inhibits orthotopic growth of human prostate cancer cells in nude mice. The purpose of this study was to determine efficacy and mechanisms of this therapy in immune-competent mice. TRAMP-C2Re3 mouse prostate cancer cells infected with 100 multiplicity of infection (MOI) of adenoviral vector encoding for mouse IFN-beta (AdmIFN-beta), but not AdE/1 (a control adenoviral vector), produced approximately 60 ng/10(5) cells/24 h of IFN-beta. The tumorigenicity of AdmIFN-beta-transduced cells was dramatically reduced in the prostates of C57BL/6 mice. A single intratumoral injection of 2 x 10(9) PFU (plaque-forming unit) of AdmIFN-beta inhibited tumor growth by 70% and prolonged survival of tumor-bearing mice. Intriguingly, this AdmIFN-beta therapy did not alter the growth of tumors in inducible nitric oxide synthase (iNOS)-null C57BL/6 mice. Immunohistochemical analysis revealed that treatment of tumors with AdmIFN-beta in wild-type C57BL/6 mice led to increased iNOS expression, decreased microvessel density, decreased cell proliferation, and increased apoptosis. Furthermore, quantitative reverse-transcriptional PCR analysis showed that AdmIFN-beta therapy, in C57BL/6 but not the iNOS-null counterparts, reduced levels of the mRNAs for angiopoietin, basic fibroblast growth factor, matrix metalloproteinase-9, transforming growth factor-beta1, vascular endothelial growth factor (VEGF)-A, and VEGF-B, as well as the antiapoptotic molecule endothelin-1. These data indicated that IFN-beta gene therapy could be effective alternative for the treatment of locally advanced prostate cancer and suggest an obligatory role of NO in IFN-beta antitumoral effects in vivo.
Collapse
Affiliation(s)
- M V Olson
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | |
Collapse
|