101
|
Oshikawa T, Okamoto M, Tano T, Uddin Ahmed S, Sasai A, Kan S, Moriya Y, Ryoma Y, Saito M, Sato M. Involvement of nitric oxide in anti-tumor effects of OK-432, a streptococcal anti-tumor immunotherapeutic agent. Int Immunopharmacol 2005; 6:764-73. [PMID: 16546707 DOI: 10.1016/j.intimp.2005.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2005] [Revised: 10/07/2005] [Accepted: 11/14/2005] [Indexed: 11/20/2022]
Abstract
We examined the role of nitric oxide (NO) induced by OK-432, a streptococcal immunotherapeutic agent, in anti-tumor effects of the OK-432 by in vitro and in vivo experiments using an NO synthase inhibitor, N-monomethyl-l-arginine acetate (NMA). The in vitro treatment of mouse splenocytes with OK-432 increased the expression of inducible NO synthase (iNOS) gene and NO production in a dose-dependent manner. Although it is well known that OK-432 induces cytokines such as interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, both of which are known to be potent NO inducers, we observed only a partial reduction of OK-432-induced NO production with the addition of anti-IFN-gamma and/or anti-TNF-alpha neutralizing antibodies. The cytotoxicity of the splenocytes increased by in vitro OK-432 stimulation was almost completely inhibited by the treatment with NMA. OK-432 administration resulted in a marked prolongation of survival and a significant inhibition of tumor growth in syngeneic tumor-bearing mice, whereas NMA significantly inhibited the anti-tumor effects of OK-432. Although the increased cytotoxicity of adherent splenocytes derived from OK-432-treated tumor-bearing mice was almost completely inhibited by NMA, only partial inhibition by NMA was observed in the cytotoxicity of the nonadherent splenocytes. These findings strongly suggest that the iNOS/NO induced by OK-432 is intimately involved in the anti-tumor effects of OK-432.
Collapse
Affiliation(s)
- Tetsuya Oshikawa
- Second Department of Oral and Maxillofacial Surgery, Tokushima University School of Dentistry, Tokushima, 7708504, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Lechner M, Lirk P, Rieder J. Inducible nitric oxide synthase (iNOS) in tumor biology: the two sides of the same coin. Semin Cancer Biol 2005; 15:277-89. [PMID: 15914026 DOI: 10.1016/j.semcancer.2005.04.004] [Citation(s) in RCA: 301] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inducible nitric oxide synthase (iNOS) is one of three key enzymes generating nitric oxide (NO) from the amino acid l-arginine. iNOS-derived NO plays an important role in numerous physiological (e.g. blood pressure regulation, wound repair and host defence mechanisms) and pathophysiological (inflammation, infection, neoplastic diseases, liver cirrhosis, diabetes) conditions. iNOS is the synthase isoform most commonly associated with malignant disease. Nevertheless, the role of iNOS during tumor development is highly complex, and incompletely understood. Both promoting and deterring actions have been described, presumably depending upon the local concentration of iNOS within the tumor microenvironment. In particular, pivotal effects such as malingnant transformation, angiogenesis, and metastasis are modulated by iNOS. On the other hand, NO derived from macrophages has a potentially cytotoxic/cytostatic effect upon tumor cells. Hence, therapeutical interference with iNOS activity is of considerable interest, especially in tumors where metastatic activity, host defence mechanisms and the level of differentiation seem to be correlated to iNOS expression. This review will aim to summarize the dual actions of iNOS as simultaneous tumor promoter and suppressor.
Collapse
Affiliation(s)
- Matthias Lechner
- Clinical Division of General and Surgical Intensive Care Medicine, Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | | |
Collapse
|
103
|
Boucquey A, Vilhardt F, Mitrovic T, Franco D, Weber A, Horellou P. Retroviral display of urokinase-binding domain fused to amphotropic envelope protein. Biochem Biophys Res Commun 2005; 331:1485-93. [PMID: 15883041 DOI: 10.1016/j.bbrc.2005.04.075] [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: 04/11/2005] [Indexed: 11/21/2022]
Abstract
Tumors frequently express urokinase (uPA) receptor (uPAR). To investigate whether uPAR can efficiently target cancerous cells using amphotropic retroviral vectors, we generated a retrovirus displaying the amino-terminal fragment (ATF) of uPA as an N-terminal extension of viral envelope protein. We also made use of a "two-step strategy" by inserting a uPA cleavage site between the ATF moiety and the envelope. We measured the ability of ATF-bearing chimeric envelopes to infect huPAR-overexpressing Madin-Darby canine kidney (MDCK) and control MDCK II cells. The ATF-viruses infected both MDCK cell lines with an equivalent efficiency, suggesting that the chimeric viruses were not sequestered by uPAR and infect cells preferentially via the Pit-2 receptor. The addition of a uPA cleavage site increased the infection level of huPAR-MDCK cells by 2-fold when uPA was present in the infection medium. Surprisingly, ATF-env viruses infected huPAR-MDCK cells 5.5-fold more efficiently in the presence of exogenous uPA. This stimulatory effect of uPA on infection of huPAR-MDCK cells by the ATF-env virus was completely abolished by methyl-beta-cyclodextrin, suggesting that this effect involves the caveolar endocytosis pathway.
Collapse
Affiliation(s)
- Antoine Boucquey
- EMI 00-20, INSERM and Université Paris XI, Bat Grégory Pincus, 80 rue du Général Leclerc, 94276 Le Kremlin Bicêtre Cedex, France
| | | | | | | | | | | |
Collapse
|
104
|
Lee KY, Jeon YJ. Macrophage activation by polysaccharide isolated from Astragalus membranaceus. Int Immunopharmacol 2005; 5:1225-33. [PMID: 15914327 DOI: 10.1016/j.intimp.2005.02.020] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2005] [Revised: 02/22/2005] [Accepted: 02/22/2005] [Indexed: 11/24/2022]
Abstract
We show that APS, a polysaccharide isolated from the roots of Astragalus membranaceus, significantly induces nitric oxide (NO) production and inducible NO synthase (iNOS) transcription through the activation of nuclear factor-kappaB/Rel (NF-kappaB/Rel). In vivo administration of APS induced NO production by peritoneal macrophages of B6C3F1 mice. APS also dose-dependently induced the production of NO in isolated mouse peritoneal macrophages and RAW 264.7, a mouse macrophage-like cell line. Moreover, iNOS protein and mRNA transcription were strongly induced by APS in RAW 264.7 cells. To further investigate the mechanism responsible for the induction of iNOS gene expression, we investigated the effect of APS 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 APS produced strong induction of NF-kappaB/Rel-dependent reporter gene expression, whereas Oct-dependent gene expression was not affected by APS. Nuclear translocation and DNA binding activity of NF-kappaB/Rel was significantly induced by APS. 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 APS stimulates macrophages to express iNOS gene through the activation of NF-kappaB/Rel.
Collapse
Affiliation(s)
- Kun Yeong Lee
- Department of Pharmacology, Chosun University College of Medicine, 375 Susukdong, Kwangju, 501-709, Republic of Korea
| | | |
Collapse
|
105
|
Le X, Wei D, Huang S, Lancaster JR, Xie K. Nitric oxide synthase II suppresses the growth and metastasis of human cancer regardless of its up-regulation of protumor factors. Proc Natl Acad Sci U S A 2005; 102:8758-63. [PMID: 15939886 PMCID: PMC1150810 DOI: 10.1073/pnas.0409581102] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Inducible nitric oxide (NO) synthase (NOS) II has been implicated in macrophage-mediated antitumor activity. However, use of the NOS II gene in cancer therapy is problematic because of the double-edged nature of NO action. Herein we show that adenoviral vectors mediated effective NOS II gene transfer into various human tumors. Production of NO significantly up-regulated multiple angiogenic molecules. However, the NO-producing tumor cells did not form tumors or metastases in ectopic or orthotopic xenograft nude mouse models. The dramatic loss of malignancy was due to NO-mediated apoptosis. We also generated a series of adenoviral vectors harboring mutant NOS II genes that expressed mutant NOS II proteins with defined levels of enzymatic activity. Tumor cells transduced with these NOS II genes produced NO at different levels, which directly correlated with the antitumor activity in vitro and in vivo. This demonstration using a relevant biological system shows that NO produces dose-dependent antitumor activity in vitro and in vivo, regardless of its up-regulation of protumor factors.
Collapse
Affiliation(s)
- Xiangdong Le
- Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, University of Texas, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
106
|
Bentrari F, Arnould L, Jackson AP, Jeannin JF, Pance A. Progesterone enhances cytokine-stimulated nitric oxide synthase II expression and cell death in human breast cancer cells. J Transl Med 2005; 85:624-32. [PMID: 15778733 DOI: 10.1038/labinvest.3700267] [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: 02/07/2023] Open
Abstract
The presence of hormone receptors is related to survival outcome in breast cancer. Previous results from our laboratory established a correlation between the presence of nitric oxide synthase II (NOSII) and nitric oxide (NO) production with progesterone receptors in a series of human breast tumours. Furthermore, this was directly related to a lower tumour grade and a lower proliferation rate of the tumour cells. To examine these results in further detail, the effect of progesterone (Pg) and 17beta-oestradiol (E2) on NOSII expression was analysed in the human breast cancer cell line MCF-7. By Northern blot and promoter activity, we show that a cytokine mix (TNF-alpha, IL-beta, and IFN-gamma) induces NOSII transcription after 6 h stimulation. In the absence of cytokines, neither hormone affects NOSII expression. However, Pg but not E2, enhances cytokine-induced NOSII transcription as well as NO synthesis, mainly by cooperation with gamma-interferon. The increase in NO accumulation in the media induced by addition of Pg to the cytokine treatment significantly increases cell death, mainly accounted for by apoptosis, as compared to the effect of cytokines alone. Our findings help clarify the role of steroid hormones in NOSII expression as well as the effect on cell viability and may suggest novel approaches towards hormonotherapy and the treatment of cancer.
Collapse
Affiliation(s)
- Fatima Bentrari
- EPHE INSERM 517, Faculty of Medicine, University of Bourgogne, Dijon, France
| | | | | | | | | |
Collapse
|
107
|
Adithi M, Nalini V, Krishnakumar S. The role of nitric oxide synthases and nitrotyrosine in retinoblastoma. Cancer 2005; 103:1701-11. [PMID: 15754329 DOI: 10.1002/cncr.20961] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND To investigate the potential involvement of the nitric oxide (NO) pathway in retinoblastoma, the authors correlated immunoreactivity for endothelial nitric oxide synthase (eNOS), inducible nitric oxide synthase (iNOS), and nitrotyrosine (NT) with the degree of tumor invasiveness in retinoblastoma. METHODS eNOS, iNOS, and NT reactivity was evaluated by immunohistochemistry in 34 archival retinoblastoma specimens and in a human Y79 retinoblastoma cell line. The tumors were divided into 2 groups: Group A tumors (n = 17 tumors) with no invasion and Group B tumors (n = 17 tumors) with invasion of the choroid, optic nerve, and/or orbit. The expression levels of eNOS, iNOS, and NT were correlated with invasiveness of the tumors. RESULTS In Group A tumors (n = 17 tumors) without invasion, eNOS was positive in 17 of 17 tumors (100%), iNOS was positive in 14 of 17 tumors (82%), and NT was positive in 17 of 17 tumors (100%). In Group B tumors (n = 17 tumors) with invasion, eNOS was positive in 17 of 17 tumors (100%), iNOS was positive in 16 of 17 tumors (94%), and NT was positive in 17 of 17 tumors (100%). The invasive cohort showed significantly higher expression of iNOS (P < 0.0001) and NT (P < 0.020) compared with the noninvasive cohort. Y79 cells also expressed eNOS, iNOS, and NT; and nonneoplastic retina was positive for eNOS, iNOS, and NT. CONCLUSIONS Taken together, the results suggested that retinoblastomas can produce NO. The roles of NO in the biology of retinoblastoma and in the prognosis for patients with retinoblastoma remain to be established.
Collapse
Affiliation(s)
- Mohan Adithi
- Department of Ocular Pathology, Vision Research Foundation, Sankara Nethralaya, Chennai, India
| | | | | |
Collapse
|
108
|
Oshikawa T, Okamoto M, Ahmed SU, Furuichi S, Tano T, Sasai A, Kan S, Kasai S, Uto Y, Nagasawa H, Hori H, Sato M. TX-1877, a bifunctional hypoxic cell radiosensitizer, enhances anticancer host response: Immune cell migration and nitric oxide production. Int J Cancer 2005; 116:571-8. [PMID: 15825172 DOI: 10.1002/ijc.21101] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We investigated in the current study the effect of TX-1877, a bifunctional hypoxic cell radiosensitizer, in augmenting anticancer host response. In the syngeneic squamous cell carcinoma-bearing mouse model, a single administration of TX-1877 significantly inhibited the primary tumor growth as well as lung metastasis. TX-1877 administration resulted in a significant infiltration of immune cells, such as CD4+T, CD8+T cells, macrophages and dendritic cells (DCs), and an increased expression of chemokines for cytotoxic T lymphocytes (CTLs), helper T-cell 1 (Th1) cells, monocytes/macrophages and DCs, in tumor tissues. Nitric oxide (NO) production and the expression of inducible NO synthase (iNOS) and interferon-gamma, a major Th1 cytokine that plays a major role in anticancer immunity, were also enhanced. Furthermore, neutralization of NO by N-monomethyl-L-arginine acetate resulted in a marked inhibition of the antitumor effect of TX-1877. In tumor-draining lymph nodes, MHC class I-restricted CD8+ memory CTLs specific for inoculated cancer cells were induced by TX-1877. In in vitro experiments, TX-1877 induced chemokines and iNOS/NO in several types of culture cells. These findings strongly suggested that TX-1877 induces migration of CD8+CTLs, CD4+Th1 cells, macrophage/monocytes and dendritic cells into the tumor site, and that this migration is mediated by chemokine induction. In addition, it was suggested that NO produced by several types of cells stimulated by TX-1877 in the tumor sites plays a major role in the anticancer effect of TX-1877. TX-1877 was thus shown to be an effective immunopotentiator as well as a hypoxic cell radiosensitizer.
Collapse
Affiliation(s)
- Tetsuya Oshikawa
- Second Department of Oral and Maxillofacial Surgery, Tokushima University School of Dentistry, 3-18-15 Kuramoto-cho, Tokushima, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Cook T, Wang Z, Alber S, Liu K, Watkins SC, Vodovotz Y, Billiar TR, Blumberg D. Nitric oxide and ionizing radiation synergistically promote apoptosis and growth inhibition of cancer by activating p53. Cancer Res 2004; 64:8015-21. [PMID: 15520210 DOI: 10.1158/0008-5472.can-04-2212] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nitric oxide (NO) is a potent tumor radiosensitizer; however, its clinical use is limited by systemic side effects. We have demonstrated previously that gene transfer of the human inducible NO synthase (iNOS) gene into tumor cells and tumors induces high-output NO production that significantly enhances tumor radioresponsiveness, with no observed side effects. Notably, iNOS gene transfer enhances tumor radioresponsiveness via apoptotic cell death. Because NO and ionizing radiation are both known to promote p53-dependent apoptosis, we hypothesized that p53 activation might be a primary mechanism for the synergy of these two genotoxic stresses. We report that NO and ionizing radiation synergistically activate p53 in colorectal cancers grown in athymic mice by augmenting phosphorylation of p53 at serine 15. The effect of NO and ionizing radiation on tumor cell apoptosis and tumor radioresponsiveness is significantly reduced in p53 knockout isogenic cancer cell lines. Furthermore, the transfer of both p53 and iNOS genes into tumor cells lacking functional p53 enhanced their radioresponsiveness more than transfer of either gene alone.
Collapse
Affiliation(s)
- Tracy Cook
- Department of Surgery and Center for Biologic Imaging, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Rao CV. Nitric oxide signaling in colon cancer chemoprevention. Mutat Res 2004; 555:107-19. [PMID: 15476855 DOI: 10.1016/j.mrfmmm.2004.05.022] [Citation(s) in RCA: 166] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 05/25/2004] [Accepted: 05/26/2004] [Indexed: 12/19/2022]
Abstract
Nitric oxide (NO) is a pleiotrophic regulator, pivotal to numerous biological processes, including vasodilation, neurotransmission, and macrophage-mediated immunity. The highly reactive free radicals, produced by NO synthases (NOS) have been implicated in the modulation of carcinogenesis. Over-expression of inducible NOS (iNOS), a common phenomenon during chronic inflammatory conditions, generates sustainable amounts of NO, that its reactive intermediates are mutagenic, causing DNA damage or impairment of DNA repair, has been well established in carcinogenesis. Recent studies also implicate NO as having a key signaling molecule that regulates processes of tumorigenesis. Increased expression of iNOS has been observed in tumors of the colon, lung, oropharynx, reproductive organs, breast, and central nervous system besides its occurrence in chronic inflammatory diseases. Progression of a large majority of human and experimental colon tumors appears to progress by NO resulting from stimulation of proinflammatory cytokines, and inactivation (nitrosylation) of p53 mediated caspase activities in the tumors, whereas in some cases it associated with induction of apoptosis and tumor regression. This dichotomy is largely explained by the complexity of signaling pathways in tumor cells, that respond to NO very differently depending on its concentration. p53 mutation, functional loss, activation, and inactivation of apoptotic proteins all have been linked with NO resistance and dependence. Evidence from both in vitro and in vivo experiments support that NO and its reactive metabolite peroxynitrite stimulate COX-2 activity leading generation of tumor growth enhancing prostaglandins. Thus, NO mediated signaling can augment the tumor growth and metastasis by promoting invasive and angiogenic properties of tumor cells, which includes triggering and activation of COX-2. Thus, developing selective inhibitors of iNOS and NO-releasing agents may lead to important strategies for chemoprevention of colon cancer. Chemoprevention studies at preclinical level with several selective inhibitors of iNOS in both chemically and transgenic models of colon cancer are encouraging.
Collapse
Affiliation(s)
- Chinthalapally V Rao
- Chemoprevention Program, American Health Foundation-Cancer Center, Institute For Cancer Prevention, Valhalla, NY 10595, USA.
| |
Collapse
|
111
|
Kim GY, Choi GS, Lee SH, Park YM. Acidic polysaccharide isolated from Phellinus linteus enhances through the up-regulation of nitric oxide and tumor necrosis factor-alpha from peritoneal macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2004; 95:69-76. [PMID: 15374609 DOI: 10.1016/j.jep.2004.06.024] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2003] [Revised: 02/13/2004] [Accepted: 06/29/2004] [Indexed: 05/08/2023]
Abstract
Medicinal mushrooms are increasingly used to treat a wide variety of disease processes. Aqueous extract from the fruiting body or mycelia of Phellinus linteus has been reported to produce antitumor and immunomodulatory activities in vivo and in vitro. However, the mechanisms underlying its tumoricidal effects are poorly understood. The tumoricidal activity of peritoneal macrophages (PM) cultured with acidic polysaccharide (PL) isolated from Phellinus linteus against B16 melanoma cells was enhanced in a dose-dependent manner; growth inhibition increased 4-fold with 200 microg/ml of PL. To further characterize the mechanisms of PL, we investigated the effects of PL on phagocytosis and the release of nitric oxide (NO), tumor necrosis factor-alpha (TNF-alpha) and reactive oxygen intermediates (ROI). To investigate the phagocytosis of PM, the uptake of Dextran (Dex)-FITC between PL-untreated and PL-treated PM was compared. We found some augment in phagocytosis of PL-treated PM compared untreated group. PL stimulated a dose-dependent increase in NO and TNF-alpha, but not in ROI production in PM. We suggested that PL has cytotoxicity against Yac-1 cells through the up-regulation of NO and TNF-alpha production. Also, PL enhanced the expression of costimulatory molecules, CD80 and CD86, and major histocompatibility complex (MHC) molecules II in PM. The ability of PL upon the up-regulation of these surface molecules involved in antigen-presenting processes may, by inference, activate T-cell-mediated immunity against malignant cells in vivo. Taken together, these results suggest that PL act as an effective immunomodulator and enhances the anti-tumoral activity of PM.
Collapse
Affiliation(s)
- Gi-Young Kim
- Department of Microbiology, Pusan National University College of Medicine, Ami-dong 1-10, Pusan 602-739, South Korea
| | | | | | | |
Collapse
|
112
|
Abstract
Arginine is a dibasic, cationic, semiessential amino acid with numerous roles in cellular metabolism. It serves as an intermediate in the urea cycle and as a precursor for protein, polyamine, creatine and nitric oxide (NO) biosynthesis. Arginine is conditionally essential since it becomes necessary under periods of growth and after recovery after injury. Arginine also promotes wound healing and functions as a secretagogue stimulating the release of growth hormone, insulin-like growth factor 1, insulin, and prolactin. Furthermore, arginine has several immunomodulatory effects such as stimulating T- and natural killer cell activity and influencing pro-inflammatory cytokine levels. The discover that l-arginine is the sole precursor for the multifunctional messenger molecule nitric oxide (NO) led to investigation into the role of arginine in numerous physiologic and pathophysiologic phenomena including cancer. Although NO was first identified in endothelial cells, it is now recognized to be generated by a variety of cell types, including several tumor cell lines and solid human tumors. Unfortunately, the precise role of NO in cancer is poorly understood but it may influence tumor initiation, promotion, and progression, tumor-cell adhesion, apoptosis angiogenesis, differentiation, chemosensitivity, radiosensitivity, and tumor-induced immunosuppression. The biological effects of NO are complex and dependent upon numerous regulatory factors. Further research is necessary to enhance our understanding of the complex mechanisms that regulate NO's role in tumor biology. A better understanding of the role of arginine-derived NO in cancer may lead to novel antineoplastic and chemopreventative strategies.
Collapse
Affiliation(s)
- D Scott Lind
- Department of Surgery, University of Florida College of Medicine, Surgical Services, North Florida South Georgia VA Health Care System, Gainesville, FL 32608, USA.
| |
Collapse
|
113
|
Thomas DD, Espey MG, Ridnour LA, Hofseth LJ, Mancardi D, Harris CC, Wink DA. Hypoxic inducible factor 1alpha, extracellular signal-regulated kinase, and p53 are regulated by distinct threshold concentrations of nitric oxide. Proc Natl Acad Sci U S A 2004; 101:8894-9. [PMID: 15178764 PMCID: PMC428443 DOI: 10.1073/pnas.0400453101] [Citation(s) in RCA: 214] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
NO produced in tumors can either positively or negatively regulate growth. To examine this dichotomy, effects of NO concentration and duration on the posttranslational regulation of several key proteins were examined in human breast MCF7 cells under aerobic conditions. We found that different concentration thresholds of NO appear to elicit a discrete set of signal transduction pathways. At low steady-state concentrations of NO (<50 nM), extracellular signal-regulated kinase (ERK) phosphorylation was induced via a guanylate cyclase-dependent mechanism. Hypoxic inducible factor 1alpha (HIF-1alpha) accumulation was associated with an intermediate amount of NO (>100 nM), whereas p53 serine 15 phosphorylation occurred at considerably higher levels (>300 nM). ERK phosphorylation was transient during NO exposure. HIF-1alpha stabilization paralleled the presence of NO, whereas p53 serine 15 phosphorylation was detected during, and persisted after, NO exposure. The dose-dependent effects of synthetic NO donors were mimicked by activated macrophages cocultured with MCF7 cells at varying ratios. ERK and HIF-1alpha activation was similar in breast cancer cell lines either mutant (MB231) or null (MB157) in p53. The stabilization of HIF-1alpha by NO was not observed with increased MCF7 cell density, demonstrating the interrelationship between NO and O(2) consumption. The findings show that concentration and duration of NO exposure are critical determinants in the regulation of tumor-related proteins.
Collapse
Affiliation(s)
- Douglas D Thomas
- Tumor Biology Section, Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
114
|
Samdani AF, Kuchner EB, Rhines L, Adamson DC, Lawson C, Tyler B, Brem H, Dawson VL, Dawson TM. Astroglia Induce Cytotoxic Effects on Brain Tumors via a Nitric Oxide-Dependent Pathway Both in Vitro and in Vivo. Neurosurgery 2004; 54:1231-7; discussion 1237-8. [PMID: 15113479 DOI: 10.1227/01.neu.0000119576.76193.b8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2003] [Accepted: 01/14/2004] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In the central nervous system, astroglia produce nitric oxide (NO) in response to cytokines. We investigated whether cytokine stimulation of astroglia could inhibit brain tumor cell growth in vitro and prolong survival in vivo via an NO-dependent pathway. METHODS Astroglia cultures were stimulated with the cytokines lipopolysaccharide and interferon-gamma and subsequently seeded with tumor cell lines. Wild-type mice and inducible NO synthase-knockout mice received in vivo cytokine stimulation followed by B16F10 murine melanoma challenge. RESULTS Our in vitro studies demonstrate that astroglia stimulated to produce NO by the addition of cytokines dose-dependently inhibit the growth of one primary rat brain tumor cell line (9L) and three primary human brain tumor cell lines (H80, U87, and U373). This inhibition of tumor cell growth is also observed in metastatic cell lines (B16F10 melanoma, Lewis lung carcinoma, and CT26 colon). Cultured astrocytes from inducible NO synthase-knockout mice, which are incapable of induction of NO, are without the enhanced tumoricidal effect. Furthermore, when C57BL/6 mice are primed to produce NO through stereotactic intracranial administration of lipopolysaccharide plus interferon-gamma and subsequently challenged with B16F10 murine melanoma, survival is significantly prolonged, with a median survival of 26 days versus 16 days in the control group (P < 0.001). The addition of an NO synthase inhibitor (N(G)-nitro-l-arginine methyl ester) decreases this beneficial effect (median survival, 21 d). CONCLUSION These findings suggest that NO may have an important role as a defense mechanism molecule against brain tumors; stimulation or modification of this mechanism may represent a new approach to the treatment of primary and metastatic brain tumors.
Collapse
Affiliation(s)
- Amer F Samdani
- Department of Neurological Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Hu DE, Dyke SOM, Moore AM, Thomsen LL, Brindle KM. Tumor Cell-Derived Nitric Oxide Is Involved in the Immune-Rejection of an Immunogenic Murine Lymphoma. Cancer Res 2004; 64:152-61. [PMID: 14729619 DOI: 10.1158/0008-5472.can-03-1807] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The roles played by host-derived nitric oxide (NO) in the growth and subsequent immune rejection of a immunogenic murine lymphoma were investigated by growing the tumor in mice in which the gene for either inducible NO synthase (iNOS) or endothelial NOS (eNOS) had been ablated. This showed that NO from tumor-infiltrating host cells had no significant effect on either tumor growth or immune rejection, although measurements of tumor nitrite levels and protein nitration showed that there had been significant NO production in the rejected tumors, in both the eNOS and iNOS knockout mice. Inhibition of both tumor and host NOS activities, with an iNOS-selective inhibitor (1400W), a nonselective NOS inhibitor [Nomega-nitro-L-arginine methyl ester (L-NAME)], or scavenging NO with a ruthenium-based scavenger, significantly delayed tumor rejection, while having no appreciable effect on tumor growth. Incubation of tumor cells with medium taken from cultured splenocytes, that had been isolated from immunized animals and activated by incubating them with irradiated tumor cells, resulted in an increase in tumor cell NOS activity and an increase in tumor cell apoptosis, which could be inhibited using L-NAME. We propose that, during the immune rejection of this tumor model, there is induction of tumor NOS activity by cytokines secreted by activated lymphocytes within the tumor and that this results in increased levels of tumor NO that induce tumor cell apoptosis and facilitate immune rejection of the tumor.
Collapse
Affiliation(s)
- De-En Hu
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| | | | | | | | | |
Collapse
|
116
|
KWON OD, YIM CY, JEONG KS, JUNG KY, McGREGOR JR, BASTIAN NR, SAMLOWSKI WE. Suppression of Cytokine-Inducible Nitric Oxide Synthesis During Intraperitoneal Meth A Tumor Growth. J Vet Med Sci 2004. [DOI: 10.1292/jvms.66.357] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Oh-Deog KWON
- University of Utah Cancer Immunotherapy Program
- Department of Internal Medicine (Oncology and Infectious Disease) of the University of Utah School of Medicine
- College of Veterinary Medicine, Kyungpook National University
| | - Chang-Yeol YIM
- Department of Internal Medicine (Hematology/Oncology) of the Chonbuk National University Medical School
| | - Kyu-Shik JEONG
- College of Veterinary Medicine, Kyungpook National University
| | - Kyu-Yong JUNG
- Department of Pharmacology, Wonkwang University School of Medicine
| | - John R. McGREGOR
- University of Utah Cancer Immunotherapy Program
- Department of Internal Medicine (Oncology and Infectious Disease) of the University of Utah School of Medicine
- Huntsman Cancer Institute
| | - Neil R. BASTIAN
- Department of Internal Medicine (Oncology and Infectious Disease) of the University of Utah School of Medicine
| | - Wolfram E. SAMLOWSKI
- University of Utah Cancer Immunotherapy Program
- Department of Internal Medicine (Oncology and Infectious Disease) of the University of Utah School of Medicine
- Huntsman Cancer Institute
| |
Collapse
|
117
|
Simeone AM, Broemeling LD, Rosenblum J, Tari AM. HER2/neu reduces the apoptotic effects of N-(4-hydroxyphenyl)retinamide (4-HPR) in breast cancer cells by decreasing nitric oxide production. Oncogene 2003; 22:6739-47. [PMID: 14555987 DOI: 10.1038/sj.onc.1206786] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
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.
Collapse
Affiliation(s)
- Ann-Marie Simeone
- Department of Bioimmunotherapy, Section of Immunobiology and Drug Carriers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
118
|
Lee KY, Jeon YJ. Polysaccharide isolated from Poria cocos sclerotium induces NF-kappaB/Rel activation and iNOS expression in murine macrophages. Int Immunopharmacol 2003; 3:1353-62. [PMID: 12946432 DOI: 10.1016/s1567-5769(03)00113-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We show that PCSC, a polysaccharide isolated from the sclerotium of Poria cocos with 1% sodium carbonate, significantly induces nitric oxide (NO) production and inducible NO synthase (iNOS) transcription through the activation of nuclear factor-kappaB/Rel (NF-kappaB/Rel). In vivo administration of PCSC induced NO production by peritoneal macrophages of B6C3F1 mice. PCSC also dose-dependently induced the production of NO in isolated mouse peritoneal macrophages and RAW 264.7, a murine macrophage-like cell line. Moreover, iNOS protein and mRNA transcription were strongly induced by PCSC in RAW 264.7 cells. To further investigate the mechanism responsible for the induction of iNOS gene expression, we investigated the effect of PCSC 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 PCSC produced strong induction of NF-kappaB/Rel-dependent reporter gene expression, whereas Oct-dependent gene expression was not affected by PCSC. DNA binding activity of NF-kappaB/Rel was significantly induced by PCSC, and this effect was mediated through the degradation of IkappaBalpha. In conclusion, we demonstrate that PCSC stimulates macrophages to express iNOS gene through the activation of NF-kappaB/Rel.
Collapse
MESH Headings
- Adjuvants, Immunologic/isolation & purification
- Adjuvants, Immunologic/pharmacology
- Animals
- Blotting, Western
- Cell Line
- Dose-Response Relationship, Drug
- Enzyme Induction
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Genes, Reporter/genetics
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred Strains
- Mycelium/chemistry
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nitric Oxide/biosynthesis
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- Polyporales/chemistry
- Polysaccharides/isolation & purification
- Polysaccharides/pharmacology
- Reverse Transcriptase Polymerase Chain Reaction
- Transcriptional Activation/drug effects
- Transfection
Collapse
Affiliation(s)
- Kun Yeong Lee
- Department of Pharmacology, Chosun University College of Medicine, 375 Susukdong, Kwangju 501-709, South Korea
| | | |
Collapse
|
119
|
Kim GY, Oh YH, Park YM. Acidic polysaccharide isolated from Phellinus linteus induces nitric oxide-mediated tumoricidal activity of macrophages through protein tyrosine kinase and protein kinase C. Biochem Biophys Res Commun 2003; 309:399-407. [PMID: 12951063 DOI: 10.1016/j.bbrc.2003.08.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mushroom polysaccharides are increasingly being utilized to treat a wide variety of diseases. Aqueous extracts from the Phellinus linteus have been reported to have anti-tumor and immunomodulatory properties. In particular, acidic polysaccharide (PL) isolated from P. linteus induced a secretory and cellular macrophage response. However, the exact mechanism by which PL regulates the macrophage functions remains unclear. PL-treated murine peritoneal macrophages in vitro and in vivo dramatically induced the production of NO. PL enhanced the lytic death of B16 cells through the production of NO. The present study examined signal molecules that may participate in PL-elicited responses by macrophages. The data demonstrated that a protein kinase C (PKC) inhibitor, staurosporine, and a protein tyrosine kinase (PTK) inhibitor, genistein, inhibited the tumoricidal activity of macrophages induced by PL. In addition, these inhibitors blocked the production of NO and the expression of surface molecules in PL-stimulated macrophages. Furthermore, CD11b/CD18 possibly mediates PL-induced cell activation. These results suggest that PL stimulates NO production for tumoricidal activity and induces cell-mediated immunity by increasing surface molecules, and the process may be a mechanism by which PL produces its therapeutic effects.
Collapse
Affiliation(s)
- Gi-Young Kim
- Department of Microbiology, College of Natural Sciences, Pusan National University, Pusan 609-735, Republic of Korea
| | | | | |
Collapse
|
120
|
Melichar B, Hu W, Patenia R, Melicharová K, Gallardo ST, Freedman R. rIFN-gamma-mediated growth suppression of platinum-sensitive and -resistant ovarian tumor cell lines not dependent upon arginase inhibition. J Transl Med 2003; 1:5. [PMID: 14572312 PMCID: PMC222988 DOI: 10.1186/1479-5876-1-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2003] [Accepted: 09/19/2003] [Indexed: 11/15/2022] Open
Abstract
Background Arginine metabolism in tumor cell lines can be influenced by various cytokines, including recombinant human interferon-γ (rIFN-γ), a cytokine that shows promising clinical activity in epithelial ovarian cancer (EOC). Methods We examined EOC cell lines for the expression of arginase in an enzymatic assay and for transcripts of arginase I and II, inducible nitric oxide synthase (iNOS), and indoleamine 2,3-dioxygenase (IDO) by reverse transcription-polymerase chain reaction. The effects of rIFN-γ on arginase activity and on tumor cell growth inhibition were determined by measuring [3H]thymidine uptake. Results Elevated arginase activity was detected in 5 of 8 tumor cell lines, and analysis at the transcriptional level showed that arginase II was involved but arginase I was not. rIFN-γ reduced arginase activity in 3 EOC cell lines but increased activity in the 2008 cell line and its platinum-resistant subline, 2008.C13. iNOS transcripts were not detected in rIFN-γ-treated or untreated cell lines. In contrast, IDO activity was induced or increased by rIFN-γ. Suppression of arginase activity by rIFN-γ in certain cell lines suggested that such inhibition might contribute to its antiproliferative effects. However, supplementation of the medium with polyamine pathway products did not interfere with the growth-inhibitory effects of rIFN-γ EOC cells. Conclusions Increased arginase activity, specifically identified with arginase II, is present in most of the tested EOC cell lines. rIFN-γ inhibits or stimulates arginase activity in certain EOC cell lines, though the decrease in arginase activity does not appear to be associated with the in vitro antiproliferative activity of rIFN-γ. Since cells within the stroma of EOC tissues could also contribute to arginine metabolism following treatment with rIFN-γ or rIFN-γ-inducers, it would be helpful to examine these effects in vivo.
Collapse
Affiliation(s)
- Bohuslav Melichar
- Department of Oncology and Radiotherapy, Charles University Medical School, Hradec Kralove, Czech Republic
| | - Wei Hu
- Department of Gynecologic Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, U.S.A
| | - Rebecca Patenia
- Department of Gynecologic Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, U.S.A
| | - Karolina Melicharová
- Department of Gerontology and Metabolic Care, Charles University Medical School, Hradec Kralove, Czech Republic
| | - Stacie T Gallardo
- Department of Gynecologic Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, U.S.A
| | - Ralph Freedman
- Department of Gynecologic Oncology, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas, U.S.A
- Program in Immunology, The University of Texas Graduate School of Biomedical Sciences at Houston, U.S.A
| |
Collapse
|
121
|
Miyazawa M, Suzuki H, Masaoka T, Kai A, Suematsu M, Nagata H, Miura S, Ishii H. Suppressed apoptosis in the inflamed gastric mucosa of Helicobacter pylori-colonized iNOS-knockout mice. Free Radic Biol Med 2003; 34:1621-30. [PMID: 12788482 DOI: 10.1016/s0891-5849(03)00218-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
UNLABELLED Deregulated cell turnover in Helicobacter pylori (H. pylori)-colonized gastric mucosa has been suggested to be linked to the gastric carcinogenesis pathway. We previously reported attenuation of apoptosis and enhancement of cellular proliferation in the H. pylori-colonized gastric mucosa of Mongolian gerbils as compared to that in mice, which might reflect a specific link between H. pylori colonization and carcinogenesis in the Mongolian gerbils; the difference between the two strains could be attributable to differences in the host genetic background. Inducible-type nitric oxide synthase (iNOS) is thought to participate in not only the inflammatory response, but also in the regulation of gastric mucosal cell turnover in H. pylori-colonized gastric mucosa. Thus, the present study was designed to examine gastric leukocyte activation and epithelial cell apoptosis in the gastric mucosa following H. pylori inoculation in iNOS-knockout mice. METHODS iNOS-knockout mice (iNOS(-/-)) and their iNOS(+/+) littermates were orally inoculated with the Sydney strain of H. pylori (SS1, 10(8) colony-forming units [CFU]). H. pylori infection was confirmed by microaerobic bacterial culture. The stomach of each mouse was evaluated 14 weeks and 30 weeks after the inoculation. Gastric mucosal accumulation of polymorphonuclear leukocytes (PMN) was assessed by determining the myeloperoxidase (MPO) activity and histological score based on the updated Sydney system. The level of apoptosis was determined by estimation of the cytoplasmic levels of mono- and oligonucleosomes and by the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling method. RESULTS The SS1-inoculated mice showed persistent H. pylori colonization for 12 weeks. While gastric mucosal PMN infiltration increased following SS1 inoculation in both iNOS(+/+) and iNOS(-/-)strains, enhanced DNA fragmentation was observed in only SS1-colonized iNOS(+/+) mice, and not in the iNOS(-/-) mice. In conclusion, although the recruitment of PMN in response to H. pylori was evoked even in the gastric mucosa of iNOS(-/-) mice, epithelial cell apoptosis induced by H. pylori was attenuated in this strain. These data suggest that iNOS may play an important role in promoting apoptosis in the H. pylori-infected inflamed gastric mucosa, and that persistent inflammation without apoptosis in iNOS(-/-) mice with H. pylori infection may be linked to preneoplastic transformation.
Collapse
Affiliation(s)
- Masaharu Miyazawa
- Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
122
|
Warke VG, Nambiar MP, Krishnan S, Tenbrock K, Geller DA, Koritschoner NP, Atkins JL, Farber DL, Tsokos GC. Transcriptional activation of the human inducible nitric-oxide synthase promoter by Kruppel-like factor 6. J Biol Chem 2003; 278:14812-9. [PMID: 12590140 DOI: 10.1074/jbc.m300787200] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nitric oxide is a ubiquitous free radical that plays a key role in a broad spectrum of signaling pathways in physiological and pathophysiological processes. We have explored the transcriptional regulation of inducible nitric-oxide synthase (iNOS) by Krüppel-like factor 6 (KLF6), an Sp1-like zinc finger transcription factor. Study of serial deletion constructs of the iNOS promoter revealed that the proximal 0.63-kb region can support a 3-6-fold reporter activity similar to that of the full-length 16-kb promoter. Within the 0.63-kb region, we identified two CACCC sites (-164 to -168 and -261 to -265) that bound KLF6 in both electrophoretic mobility shift and chromatin immunoprecipitation assays. Mutation of both these sites abrogated the KLF6-induced enhancement of the 0.63-kb iNOS promoter activity. The binding of KLF6 to the iNOS promoter was significantly increased in Jurkat cells, primary T lymphocytes, and COS-7 cells subjected to NaCN-induced hypoxia, heat shock, serum starvation, and phorbol 12-myristate 13-acetate/ ionophore stimulation. Furthermore, in KLF6-transfected and NaCN-treated COS-7 cells, there was a 3-4-fold increase in the expression of the endogenous iNOS mRNA and protein that correlated with increased production of nitric oxide. These findings indicate that KLF6 is a potential transactivator of the human iNOS promoter in diverse pathophysiological conditions.
Collapse
Affiliation(s)
- Vishal G Warke
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
The presence of activated oncogenes and/or inactivated tumor suppressor genes may result in constitutive activation of multiple transcription factors. This may be especially true in the early stages of tumor development. At advanced stages, however, uncontrolled tumor growth and the consequent development of a stress microenvironment, such as hypoxia, acidosis, and free radical overproduction, may further alter the activity of these transcription factors. Abnormal activation of and interplay between these factors lead to aberrant expression of multiple metastasis-related proteins and confer a tremendous survival and growth advantage to emerging metastatic variants. Understanding the expression and regulation of these molecules may shed more light on the biology of cancer metastasis as well as suggest new preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Keping Xie
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | |
Collapse
|
124
|
Wang B, Wei D, Crum VE, Richardson EL, Xiong HH, Luo Y, Huang S, Abbruzzese JL, Xie K. A novel model system for studying the double-edged roles of nitric oxide production in pancreatic cancer growth and metastasis. Oncogene 2003; 22:1771-82. [PMID: 12660813 DOI: 10.1038/sj.onc.1206386] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the present study, a model system for studying the role of nitric oxide (NO) in tumor growth and metastasis was reported. Incubation of Panc02 murine pancreatic adenocarcinoma cells in vitro with cytokines and interferon led to heterogeneous expression of NO synthase II (NOS II) protein. Clonal sublines expressing different levels of NOS II were then established using a limited dilution technique. After orthotopical implantation into the pancreas of syngeneic C57BL/6 mice, clones with a low level of NOS II expression produced tumors in pancreas, metastasized to the liver, and formed ascites, whereas those having a high level of NOS II expression did not. Liver-metastasis variants having low to high metastatic ability were also established using in vivo/in vitro passage. Compared with parental Panc02 cells exhibiting a high level of NOS II expression, these variants had a decreased level of NOS II expression. Furthermore, the heterogeneous Panc02 cells were injected intravenously into a large number of syngeneic mice. Variants that metastasized to the liver, lung, skin, peritoneum, ovary, and lymph nodes were established. All of the metastatic variants exhibited a lower level of NOS II expression than the parental Panc02 cell line did. However, the phenotypes of NOS II induction and metastatic ability were unstable. Multiple in vitro/in vivo selection led to stable low NOS II expression and high metastatic potential. Finally, to further confirm the role of NOS II expression derived from tumor cells in metastasis, poorly metastatic Panc02-H0 and highly metastatic Panc02-H7 cells were injected into the pancreas of syngeneic NOS II(-/-) mice, and groups of mice received i.p. injections of either phosphate-buffered saline or L-N(6)-(1-iminoethyl) lysine. Inhibition of NOS II activity in vivo significantly promoted distant liver metastasis. Collectively, these data show that NOS II expression is highly heterogeneous and dynamically regulated, which can directly influence tumor growth and metastasis.
Collapse
Affiliation(s)
- Bailiang Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Cianchi F, Cortesini C, Fantappiè O, Messerini L, Schiavone N, Vannacci A, Nistri S, Sardi I, Baroni G, Marzocca C, Perna F, Mazzanti R, Bechi P, Masini E. Inducible nitric oxide synthase expression in human colorectal cancer: correlation with tumor angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:793-801. [PMID: 12598314 PMCID: PMC1868089 DOI: 10.1016/s0002-9440(10)63876-x] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
To investigate the potential involvement of the nitric oxide (NO) pathway in colorectal carcinogenesis, we correlated the expression and the activity of inducible nitric oxide synthase (iNOS) with the degree of tumor angiogenesis in human colorectal cancer. Tumor samples and adjacent normal mucosa were obtained from 46 surgical specimens. Immunohistochemical expression of iNOS, vascular endothelial growth factor (VEGF), and CD31 was analyzed on paraffin-embedded tissue sections. iNOS activity and cyclic GMP levels were assessed by specific biochemical assays. iNOS protein expression was determined by Western blot analysis. iNOS and VEGF mRNA levels were evaluated using Northern blot analysis. Both iNOS and VEGF expressions correlated significantly with intratumor microvessel density (r(s) = 0.31, P = 0.02 and r(s) = 0.67, P < 0.0001, respectively). A significant correlation was also found between iNOS and VEGF expression (P = 0.001). iNOS activity and cyclic GMP production were significantly higher in the cancer specimens than in the normal mucosa (P < 0.0001 and P < 0.0001, respectively), as well as in metastatic tumors than in nonmetastatic ones (P = 0.002 and P = 0.04, respectively). Western and Northern blot analyses confirmed the up-regulation of the iNOS protein and gene in the tumor specimens as compared with normal mucosa. NO seems to play a role in colorectal cancer growth by promoting tumor angiogenesis.
Collapse
Affiliation(s)
- Fabio Cianchi
- Department of General Surgery, Medical School, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
126
|
Looms D, Tritsaris K, Pedersen AM, Nauntofte B, Dissing S. Nitric oxide signalling in salivary glands. J Oral Pathol Med 2002; 31:569-84. [PMID: 12406302 DOI: 10.1034/j.1600-0714.2002.00047.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Nitric oxide (NO) plays multiple roles in both intracellular and extracellular signalling mechanisms with implications for health and disease. This review focuses on the role of NO signalling in salivary secretion. Attention will be paid primarily to endogenous NO production in acinar cells resulting from specific receptor stimulation and to NO-regulated Ca2+ homeostasis. Due to the fact that NO readily crosses membranes by simple diffusion, endogenous NO may play a physiological role in processes as diverse as modifying the secretory output, controlling blood supply to the gland, modulating transmitter output from nerve endings, participating in the host defence barrier, and affecting growth and differentiation of surrounding tissue. Furthermore, the role of NO in the pathogenesis of human oral diseases will be considered.
Collapse
Affiliation(s)
- Dagnia Looms
- Department of Medical Physiology, Faculty of Health Sciences, Panum Institute, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
127
|
Porta C, Rizzo V, Zimatore M, Sartore-Bianchi A, Danova M, Mutti L. Intrapleural interleukin-2 induces nitric oxide production in pleural effusions from malignant mesothelioma: a possible mechanism of interleukin-2-mediated cytotoxicity? Lung Cancer 2002; 38:159-62. [PMID: 12399127 DOI: 10.1016/s0169-5002(02)00187-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Due to the frequent use of intrapleural interleukin-2 (IL-2) to treat pleural effusions from malignant mesothelioma (MMe), we measured nitric oxide (NO) end product nitrite (NO(2)(-)) in pleural effusions of 12 MMe patients with chronic or chronic-relapsing pleurisy. Through high performance liquid chromatography analysis, NO(2)(-) was found in the initial pleural fluid sample of all patients (156.25 pmol ml(-1)), and increased significantly following IL-2 intrapleural instillation, both at 24 (589.91 pmol ml(-1), P < or = 0.0005) and 48 h (756 pmol ml(-1), P< or = 0.0005). Even though it is difficult to argue if the large amounts of NO end product NO(2)(-) we observed is produced by IL-2-stimulated and recruited immune cells, by MMe cells themselves, or by both, it is possible that NO could contribute to the complex antitumor activity of IL-2.
Collapse
Affiliation(s)
- Camillo Porta
- Internal Medicine and Medical Oncology, IRCCS San Matteo University Hospital, I-27100 Pavia, Italy.
| | | | | | | | | | | |
Collapse
|
128
|
Harmey JH, Bucana CD, Lu W, Byrne AM, McDonnell S, Lynch C, Bouchier-Hayes D, Dong Z. Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer 2002; 101:415-22. [PMID: 12216068 DOI: 10.1002/ijc.10632] [Citation(s) in RCA: 196] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endotoxin/lipopolysaccharide (LPS), a cell wall component of Gram-negative bacteria, is a potent inflammatory stimulus. We previously reported that LPS increased the growth of experimental metastases in a murine tumor model. Here, we examined the effect of LPS exposure on key determinants of metastasis-angiogenesis, tumor cell invasion, vascular permeability, nitric oxide synthase (NOS) and matrix metalloproteinase 2 (MMP2) expression. BALB/c mice bearing 4T1 lung metastases were given an intraperitoneal (i.p.) injection of 10 microg LPS or saline. LPS exposure resulted in increased lung weight and incidence of pleural lesions. LPS increased angiogenesis both in vivo and in vitro. Vascular permeability in lung tissue was increased 18 hr after LPS injection. LPS increased inducible nitric oxide synthase (iNOS) and MMP2 expression in lung tumor nodules. 4T1 cells transfected with green fluorescent protein (4T1-GFP) were injected via lateral tail vein. LPS exposure resulted in increased numbers of 4T1-GFP cells in mouse lung tissue compared to saline controls, an effect blocked by the competitive NOS inhibitor, N(G) methyl-L-arginine (NMA). LPS-induced growth and metastasis of 4T1 experimental lung metastases is associated with increased angiogenesis, vascular permeability and tumor cell invasion/migration with iNOS expression implicated in LPS-induced metastasis.
Collapse
Affiliation(s)
- Judith H Harmey
- Department of Cell Biology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Kostourou V, Robinson SP, Cartwright JE, Whitley GSJ. Dimethylarginine dimethylaminohydrolase I enhances tumour growth and angiogenesis. Br J Cancer 2002; 87:673-80. [PMID: 12237779 PMCID: PMC2364234 DOI: 10.1038/sj.bjc.6600518] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2002] [Revised: 04/05/2002] [Accepted: 06/26/2002] [Indexed: 11/17/2022] Open
Abstract
Angiogenesis is a prerequisite for tumour progression and is highly regulated by growth factors and cytokines a number of which also stimulate the production of nitric oxide. Asymmetric dimethylarginine is an endogenous inhibitor of nitric oxide synthesis. Asymmetric dimethylarginine is metabolised by dimethylarginine dimethylaminohydrolase. To study the effect of dimethylarginine dimethylaminohydrolase on tumour growth and vascular development, the rat C6 glioma cell line was manipulated to overexpress the rat gene for dimethylarginine dimethylaminohydrolase I. Enhanced expression of dimethylarginine dimethylaminohydrolase I increased nitric oxide synthesis (as indicated by a two-fold increase in the production of cGMP), expression and secretion of vascular endothelial cell growth factor, and induced angiogenesis in vitro. Tumours derived from these cells grew more rapidly in vivo than cells with normal dimethylarginine dimethylaminohydrolase I expression. Immunohistochemical and magnetic resonance imaging measurements were consistent with increased tumour vascular development. Furthermore, dimethylarginine dimethylaminohydrolase activity was detected in a series of human tumours. This data demonstrates that dimethylarginine dimethylaminohydrolase plays a pivotal role in tumour growth and the development of the tumour vasculature by regulating the concentration of nitric oxide and altering vascular endothelial cell growth factor production.
Collapse
Affiliation(s)
- V Kostourou
- Department of Biochemistry and Immunology, St George's Hospital Medical School, London SW17 0RE, UK
| | | | | | | |
Collapse
|
130
|
Yamazaki S, Morita T, Endo H, Hamamoto T, Baba M, Joichi Y, Kaneko S, Okada Y, Okuyama T, Nishino H, Tokue A. Isoliquiritigenin suppresses pulmonary metastasis of mouse renal cell carcinoma. Cancer Lett 2002; 183:23-30. [PMID: 12049811 DOI: 10.1016/s0304-3835(02)00113-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Isoliquiritigenin is a chalcone isolated from licorice and shallots. The ability of isoliquiritigenin to suppress metastasis was examined in a pulmonary metastasis model of mouse renal cell carcinoma. Isoliquiritigenin significantly reduced pulmonary metastasis, without any weight loss or leukocytopenia. Isoliquiritigenin suppressed in vitro proliferation of carcinoma cells, potentiated nitric oxide production by lipopolysaccharide-stimulated macrophages, and facilitated cytotoxicity of splenic lymphocytes in vitro. These findings suggest activation of macrophages, activation of cytotoxicity of lymphocytes, and direct cytotoxicity as possible mechanisms of metastasis suppression by isoliquiritigenin. In addition, isoliquiritigenin prevented severe leukocytopenia caused by administration of 5-fluorouracil.
Collapse
Affiliation(s)
- Shoji Yamazaki
- Department of Hospital Pharmacy, Jichi Medical School, 3311 Yakushiji, Minamikawachi, Tochigi 329-0498, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Millet A, Bettaieb A, Renaud F, Prevotat L, Hammann A, Solary E, Mignotte B, Jeannin JF. Influence of the nitric oxide donor glyceryl trinitrate on apoptotic pathways in human colon cancer cells. Gastroenterology 2002; 123:235-46. [PMID: 12105852 DOI: 10.1053/gast.2002.34310] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS We have previously reported the role of nitric oxide in colon tumor regression in vivo. The present study was designed to explore the influence of an endogenous nitric oxide donor, glyceryl trinitrate (GTN), on cell death pathways in colon cancer cells. METHODS Human colon cancer cell lines were treated with the NO donor GTN. Apoptosis was identified by morphological criteria and the terminal deoxynucleotidyl transferase-mediated deoxyuridine (TUNEL) method. The mitochondrial transmembrane potential was studied by flow cytometry, cytochrome c release by Western blot, and caspase activation by combining fluorogenic peptide substrates, peptide inhibitors, and immunoblotting. Expression of death receptors was studied by flow cytometry and confocal microscopy. RESULTS GTN induces a dose- and time-dependent cell death by apoptosis in colon cancer cells. This cell death pathway involves the mitochondria and caspases, mainly caspase-1 and caspase-10. In contrast, caspase-3 activation is a late and limited event. Death receptors are not involved in GTN-mediated cell death, while GTN sensitizes tumor cells to Fas-ligand-induced apoptosis. This permissive effect correlates with an increased expression of Fas receptor and a decreased expression of several endogenous inhibitors of apoptosis (IAPs). CONCLUSIONS Our results indicate that GTN (1) activates an unusual caspase cascade to induce apoptosis in colon cancer cells and (2) sensitizes these cells to Fas-mediated cell death by increasing the expression of Fas and decreasing the expression of several IAPs.
Collapse
Affiliation(s)
- Anne Millet
- Cancer Immunotherapy Laboratory of the Ecole Pratique des Hautes Etudes, INSERM U-517, Faculties of Medicine and Pharmacy, and CHU Le Bocage, Dijon, France
| | | | | | | | | | | | | | | |
Collapse
|
132
|
Ohno T, Inoue M, Ogihara Y. Suppressive effect of Shichimotsu-koka-to (Kampo medicine) on pulmonary metastasis of B16 melanoma cells. Biol Pharm Bull 2002; 25:880-4. [PMID: 12132662 DOI: 10.1248/bpb.25.880] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Shichimotsu-koka-to (SKT) is a Kampo (traditional Japanese herbal) medicine, which is used in Japan to treat hypertension and atherosclerosis. We investigated the inhibitory effect of SKT on experimental pulmonary metastasis of B16 melanoma cells. The intake of SKT at a dose of 430 mg/kg for 6 weeks from 2 weeks before tumor inoculation significantly reduced the number of metastatic surface nodules in the lung and extended the life span. When the duration of SKT intake was examined, survival time was not affected by preintake before B16 melanoma cell inoculation and was slightly extended by postintake after B16 melanoma cell inoculation, although the life span was prolonged by intake throughout the experiment. To address the mechanism underlying the antimetastatic effect of SKT, we studied whether SKT modulated macrophage function, which is involved in killing tumor cells. The intake of SKT for 6 weeks dose dependently increased nitric oxide (NO) production by macrophages following stimulation with lipopolysaccharide. The elevated NO was found to serve as a cytotoxic mediator against B16 melanoma cells in co-culture with macrophages. On the contrary, B16 melanoma-conditioned medium reduced NO production by macrophages. However, SKT treatment reversed the reduction in NO production by the conditioned medium significantly. These findings may suggest that macrophage function-modulating activity by SKT appears to underlie its antimetastatic activity, which leads to a decrease in the number of lung metastatic surface nodules and the extension of life span.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Cell Survival/drug effects
- Culture Media, Conditioned
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Lung Neoplasms/drug therapy
- Lung Neoplasms/secondary
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/metabolism
- Male
- Medicine, Kampo
- Melanoma, Experimental/secondary
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Nitric Oxide/biosynthesis
- Pharmaceutical Preparations/administration & dosage
- Plant Extracts/administration & dosage
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Takamasa Ohno
- Department of Pharmacognosy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | | | | |
Collapse
|
133
|
Kopetz ES, Nelson JB, Carducci MA. Endothelin-1 as a target for therapeutic intervention in prostate cancer. Invest New Drugs 2002; 20:173-82. [PMID: 12099577 DOI: 10.1023/a:1015630513908] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The endothelins, a family of potent vasoconstricting peptides, have been implicated in the pathophysiology of advanced prostate cancer. Two endothelin receptors, ET-A and ET-B are found in normal prostate tissue. Malignant prostate cells are notable for the loss of ET-B receptors and increased levels of endothelin-1 [ET-1]; this distortion of the endothelin system may be a significant factor in the progression of prostate cancer. Proposed roles for endothelin in prostate cancer include growth promotion, apoptosis inhibition, bone formation, and stimulation of nociceptive receptors. ET-1 can act alone as a mitogen, but its effects are greatest as a comitogen with a variety of growth factors, including basic fibroblast growth factor, insulin-like growth factors, and platelet derived growth factor. Although their exact functions are unclear, ET-1, in conjunction with vascular endothelial growth factor, appears to play a major role in tumor angiogenesis. By a variety of methods, ET-1 alters the balance of osteoblast and osteoclasts to the favor new bone formation that is characteristic of metastatic disease. Several studies indicate that the refractory pain of metastatic cancer is related to the direct nociceptive effects ET-1. These findings suggest that ET receptors are promising therapeutic targets for pharmacologic intervention. Early clinical trials indicate that the ET-A receptor antagonist used in prostate cancer is reasonably well tolerated with mild but pervasive symptoms related to ET-1's vasoconstrictive effects. Results of ongoing clinical trials are eagerly awaited in order to see if the hypothetical promise of ET antagonism will result in clinical success.
Collapse
Affiliation(s)
- E Scott Kopetz
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
134
|
Cuny E, Loiseau H, Penchet G, Ellie E, Arsaut J, Vital A, Vincendeau P, Demotes-Mainard J. Association of elevated glial expression of interleukin-1beta with improved survival in patients with glioblastomas multiforme. J Neurosurg 2002; 96:294-301. [PMID: 11838804 DOI: 10.3171/jns.2002.96.2.0294] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The aim of this study was to investigate the association of interleukin-1beta (IL-1beta) expression with improved survival in patients with glioblastomas multiforme (GBMs). Immune and vascular host-tumor interactions play a pivotal role in the control of tumor development, and inflammatory mechanisms may participate in the host's defense against tumor cells. Expression of proinflammatory cytokines and of inducible nitric oxide synthase (iNOS) has been noted in various types of malignant tumors, raising the possibility that endogenous expression of cytokines and the resulting cytotoxic action of sustained NO production play a role in the control of tumor growth. Indeed, human GBMs express variable amounts of iNOS. METHODS In this study, the expression of iNOS and of cytokines known to upregulate IL-1beta, tumor necrosis factor-alpha, interferon-gamma or downregulate iNOS transcription (IL-10, transforming growth factor [TGF]beta1, and TGFbeta2) were measured using reverse transcription-polymerase chain reaction with competitor DNA in 39 samples of human GBM. The iNOS level in GBM was positively correlated with IL-1beta messenger (m)RNA, but not with the other cytokines tested. Immunocytochemical double labeling revealed that both anti-iNOS immunoreactivity and anti-IL-1beta immunoreactivity colocalized with glial fibrillary acidic protein immunoreactivity in GBM. Some macrophage/microglial cells also expressed iNOS, but not IL-1beta. Comparison of biological data with clinical parameters indicated that the survival duration was enhanced when levels of IL-1beta mRNA were elevated or when levels of TGFbeta2 were low, but was independent of the level of iNOS mRNA within the tumor. CONCLUSIONS Taken together, these data indicate that the proinflammatory cytokine IL-1beta produced within GBM by glial-derived cells has a negative impact on tumor growth through a mechanism independent of iNOS induction.
Collapse
Affiliation(s)
- Emmanuel Cuny
- INSERM U-394 Neurobiologie Intégrative, Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
135
|
Abstract
Macrophages can kill tumor cells by releasing high levels of nitric oxide (NO) and related reactive nitrogen species such as nitroxyl and peroxynitrite, after up-regulation of expression of the inducible nitric oxide synthase gene (iNOS). In this paper we describe two novel human cell lines that are capable of expressing high levels of iNOS under the control of analogues of either the insect hormone ecdysone or tetracycline. We have entrapped these iNOS-expressing cells within a semipermeable alginate-poly-L-lysine membrane as a means of delivery to tumor sites in a nude mouse model. These encapsulated cells can be induced to generate sustainable high concentrations NO and reactive nitrogen species at tumor sites after treatment either with ponasterone A or muristerone A or with doxycycline. Delivery of these iNOS-expressing cells to tumors formed from human ovarian cancer SKOV-3 cells results in 100% killing, whereas treatment of tumors formed from human colon cancer DLD-1 cells results in 54% killing. We show that in these iNOS-expressing cells, tumor killing is associated with concomitant up-regulation of the Fas/FasL proteins.
Collapse
Affiliation(s)
- Weiming Xu
- The Wolfson Institute for Biomedical Research, UCL, London, WC1E 6BT, UK
| | | | | |
Collapse
|
136
|
Chen YK, Hsue SS, Lin LM. The mRNA expression of inducible nitric oxide synthase in DMBA-induced hamster buccal-pouch carcinomas using reverse transcription-polymerase chain reaction. J Oral Pathol Med 2002; 31:82-6. [PMID: 11896828 DOI: 10.1046/j.0904-2512.2001.00034.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Three isoforms of NO synthase (NOS) have been identified: endothelial NOS, neuronal NOS, and inducible NOS (iNOS). The enhanced expression of iNOS, at the protein level, has been reported previously in certain chemically induced oral carcinomas in hamster buccal-pouch mucosa, however, the corresponding expression of iNOS, at the mRNA level, has not yet been demonstrated. The purpose of the present study is to assess the iNOS mRNA expression level in 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal-pouch carcinomas using a reverse transcription-polymerase chain reaction (RT-PCR). METHODS Thirty-three outbred, young (six-weeks old), male, Syrian golden hamsters (Mesocricetus auratus) were randomly divided into one experimental group (13 animals) and two control groups (10 animals each). The pouches of a group of 13 animals of the experimental group were bilaterally painted with a 0.5% DMBA solution three times a week for 12 weeks. Each animal of one control group was similarly treated with mineral oil only, while the other control group of 10 animals remained untreated throughout the experiment. RESULTS Areas of dysplasia and squamous-cell carcinomas, with a 100% tumor incidence, developed for all of the DMBA-treated buccal pouches. The mineral oil-treated and untreated pouches had no obvious changes. A band of 499-bp, corresponding to iNOS mRNA, was present in all the DMBA-treated hamster buccal-pouch mucosa animals, but not in the untreated animals or the animals treated with mineral oil. Upon direct sequencing, this 499-bp band was confirmed to be part of the iNOS gene. CONCLUSIONS This study demonstrated that increased iNOS mRNA expression could contribute to the mechanism for experimentally induced oral carcinogenesis.
Collapse
Affiliation(s)
- Yuk-Kwan Chen
- Oral Pathology Department, School of Dentistry, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | | | | |
Collapse
|
137
|
Abstract
Nitric oxide (NO), an important molecule involved in neurotransmission, vascular homeostasis, immune regulation, and host defense, is generated from a guanido nitrogen of L-arginine by the family of NO synthase enzymes. Large amounts of NO produced for relatively long periods of time (days to weeks) by inducible NO synthase in macrophages and vascular endothelial cells after challenge with lipopolysaccharide or cytokines (such as interferons, tumor necrosis factor-alpha, and interleukin-1), are cytotoxic for various pathogens and tumor cells. This cytotoxic effect against tumor cells was found to be associated with apoptosis (programmed cell death). The mechanism of NO-mediated apoptosis involves accumulation of the tumor suppressor protein p53, damage of different mitochondrial functions, alterations in the expression of members of the Bcl-2 family, activation of the caspase cascade, and DNA fragmentation. Depending on the amount, duration, and the site of NO production, this molecule may not only mediate apoptosis in target cells but also protect cells from apoptosis induced by other apoptotic stimuli. In this review, we will concentrate on the current knowledge about the role of NO as an effector of apoptosis in tumor cells and discuss the mechanisms of NO-mediated apoptosis.
Collapse
Affiliation(s)
- V Umansky
- Division of Cellular Immunology, Tumor Immunology Program, German Cancer Research Center, D-69120 Heidelberg, Germany.
| | | |
Collapse
|
138
|
You HJ, Choi CY, Kim JY, Park SJ, Hahm KS, Jeong HG. Ursolic acid enhances nitric oxide and tumor necrosis factor-alpha production via nuclear factor-kappaB activation in the resting macrophages. FEBS Lett 2001; 509:156-60. [PMID: 11741581 DOI: 10.1016/s0014-5793(01)03161-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ursolic acid (UA), a pentacyclic triterpene acid, is reported to have anti-tumor activities; however, the mechanism underlying its anti-tumorigenic effects is poorly understood. To further determine the mechanism of UA, we investigated the effects of UA on the release of nitric oxide (NO) and tumor necrosis factor-alpha (TNF-alpha), and on the level of inducible nitric oxide synthase (iNOS) and TNF-alpha gene expression in mouse resting macrophages. We found that UA elicited a dose-dependent increase in NO and TNF-alpha production, and the level of iNOS and TNF-alpha mRNA. Transient expression and electrophoretic mobility shift assays with nuclear factor-kappaB (NF-kappaB) binding sites revealed that the increased level of iNOS mRNA and TNF-alpha mRNA induced by UA were mediated by the NF-kappaB transcription factor complex. These results demonstrate that UA stimulates NO and TNF-alpha release and is able to upregulate iNOS and TNF-alpha expression through NF-kappaB transactivation in the resting macrophages.
Collapse
Affiliation(s)
- H J You
- Department of Pharmacy, College of Pharmacy, Chosun University, Kwangju, South Korea
| | | | | | | | | | | |
Collapse
|
139
|
Azizzadeh B, Yip HT, Blackwell KE, Horvath S, Calcaterra TC, Buga GM, Ignarro LJ, Wang MB. Nitric oxide improves cisplatin cytotoxicity in head and neck squamous cell carcinoma. Laryngoscope 2001; 111:1896-900. [PMID: 11801965 DOI: 10.1097/00005537-200111000-00004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To test whether nitric oxide (NO) enhances the cytotoxicity of cisplatin in a head and neck squamous cell carcinoma (HNSCC) cell line. BACKGROUND Cisplatin is one of the most frequently used chemotherapeutic agents in the treatment of HNSCC. NO has been shown to play an important role in regulating tumor growth. Previous studies demonstrate that NO can enhance the cytotoxicity of cisplatin in Chinese hamster lung fibroblasts. In this report, we examined the in vitro interaction of NO and cisplatin in a HNSCC cell line. MATERIALS AND METHODS CCL23 cells were pretreated with three different NO donors: PAPA/NO (t 1/2 = 15 min), DPTA/NO (t 1/2 = 3 h), and DETA/NO (t 1/2 = 20 h). The cells were rinsed and exposed for 6 hours to a culture medium containing cisplatin. Cell survival and LD50 of cisplatin were calculated with and without NO pretreatment. RESULTS PAPA/NO and DPTA/NO did not show any cytotoxic activity and did not change the LD50 of cisplatin. DETA/NO when used alone resulted in 25.6% cell death at its peak dose (100 microM). Pretreatment with DETA/NO resulted in almost a threefold reduction of the LD50 of cisplatin (6.8 vs. 2.4 microg/mL). Pretreatment with DETA/NO sensitized the HNSCC cells to subsequent cisplatin activity (two-sided P =.00016). CONCLUSION Pretreatment of HNSCC cells with long-acting NO donors enhances cisplatin activity. Short- and medium-acting NO donors do not exert a toxic effect and do not augment the activity of cisplatin. NO agonists should be considered in the future as a possible adjunct to cisplatin in the treatment of HNSCC. Further studies with animal models are necessary to further clarify this relationship.
Collapse
Affiliation(s)
- B Azizzadeh
- Division of Head and Neck Surgery, Department of Molecular and Medical Pharmacology, University of California Los Angeles School of Medicine, Los Angeles, California 90095-1624, USA
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Choi CY, You HJ, Jeong HG. Nitric oxide and tumor necrosis factor-alpha production by oleanolic acid via nuclear factor-kappaB activation in macrophages. Biochem Biophys Res Commun 2001; 288:49-55. [PMID: 11594750 DOI: 10.1006/bbrc.2001.5727] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Oleanolic acid (OA), a pentacyclic triterpene acid, is reported to have antitumor activities; however, the mechanism underlying its antitumorigenic effects is poorly understood. To further determine the mechanism of OA, we investigated the effects of OA on the release of nitric oxide (NO) and tumor necrosis factor-alpha (TNF-alpha) and on the level of inducible nitric oxide synthase (iNOS) and TNF-alpha gene expression in mouse macrophages. We found that OA elicited a dose-dependent increase in NO and TNF-alpha production. Reverse transcription-polymerase chain reaction showed that the increased NO and TNF-alpha secretion were due to an increase in iNOS mRNA and TNF-alpha mRNA, respectively. Since iNOS and TNF-alpha transcription have recently been shown to be under the control of the NF-kappaB transcription factor, the effects of OA on NF-kappaB activation were examined using a transient transfection assay and an electrophoretic mobility shift assay (EMSA). Transient expression assays with NF-kappaB binding sites linked to the luciferase gene revealed that the increased levels of iNOS mRNA and TNF-alpha mRNA induced by OA were mediated by the NF-kappaB transcription factor complex. Using DNA fragments containing the NF-kappaB binding sequence, OA was shown to activate the protein/DNA binding of NF-kappaB to its cognate site as measured by EMSA. These results demonstrate that OA stimulates NO and TNF-alpha release and is able to upregulate iNOS and TNF-alpha expression through NF-kappaB transactivation, which may be the mechanism whereby OA elicits its biological effects.
Collapse
Affiliation(s)
- C Y Choi
- Department of Pharmacy, Chosun University, 375 Seosuk-dong, Kwangju 501-759, South Korea
| | | | | |
Collapse
|
141
|
Scott DJ, Hull MA, Cartwright EJ, Lam WK, Tisbury A, Poulsom R, Markham AF, Bonifer C, Coletta PL. Lack of inducible nitric oxide synthase promotes intestinal tumorigenesis in the Apc(Min/+) mouse. Gastroenterology 2001; 121:889-99. [PMID: 11606502 DOI: 10.1053/gast.2001.27994] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS The role of the inducible isoform of nitric oxide synthase (Nos2 or iNOS) in intestinal tumorigenesis is unclear. Conflicting data also exist regarding the ability of Nos2 to modulate expression and/or activity of cyclooxygenase 2 (Cox-2), which promotes intestinal tumorigenesis. Therefore, we determined the effect of a null Nos2 genotype on intestinal tumorigenesis and Cox-2 expression/activity in the Apc(Min/+) mouse model of familial adenomatous polyposis. METHODS Apc(Min/+)Nos2(-/-) mice were generated by successive crosses between C57BL/6-Apc(Min/+) and C57BL/6-Nos2(tm1Lau) mice. Adenoma characteristics of age-matched Apc(Min/+)Nos2(+/+) and Apc(Min/+)Nos2(-/-) mice were compared. The level and cellular localization of Nos2 messenger RNA (mRNA) expression in Apc(Min/+)Nos2(+/+) mouse intestine was determined. Cox-2 expression and activity were measured in both intestinal tissue and bone marrow-derived macrophages in vitro. RESULTS Apc(Min/+)Nos2(-/-) mice developed significantly more intestinal adenomas than Apc(Min/+)Nos2(+/+) littermates. Epithelial cell Nos2 mRNA expression was decreased in adenomas compared with histologically normal Apc(Min/+)Nos2(+/+) intestine. There was no significant difference in Cox-2 expression or activity in either intestine or bone marrow-derived macrophages from Apc(Min/+)Nos2(+/+) and Apc(Min/+)Nos2(-/-) animals. CONCLUSIONS Nos2 plays an antineoplastic role in the Apc(Min/+) mouse model of familial adenomatous polyposis. Nos2 does not modulate Cox-2 expression or activity in the Apc(Min/+) mouse.
Collapse
Affiliation(s)
- D J Scott
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds, England, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Mitsumoto A, Kim KR, Oshima G, Nakagawa Y. Inhibitory effects of S-nitrosoglutathione on cell proliferation and DNA synthesis: possible role of glyoxalase I inactivation. Chem Biol Interact 2001; 137:105-21. [PMID: 11551528 DOI: 10.1016/s0009-2797(01)00224-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We investigated the inhibitory effects of S-nitrosoglutathione (GSNO) on cell proliferation, DNA synthesis and several enzymatic activities using spontaneously immortalized human endothelial cells (ECV304). Proliferation of ECV304 was inhibited by GSNO in a dose-dependent manner (125-1000 microM). DNA synthesis was decreased 2 h after addition of GSNO to cells and was markedly repressed from 20 h after the addition. The activity of ribonucleotide reductase, a rate-limiting enzyme for DNA synthesis, was unchanged in GSNO-treated cells. GSNO inhibited less than 40% of mitochondrial respiration activity, and the membrane potential and cellular levels of ATP were not significantly decreased by GSNO. GSNO had no inhibitory effect on activities of glutathione peroxidase, glutathione S-transferase and glutathione reductase. However, glyoxalase I (Glo I) activity was decreased to 20% of the control level within 60 min, and was consistently repressed during exposure to GSNO for 20 h. A membrane-permeable Glo I inhibitor, S-bromobenzylglutathione diethylester, inhibited proliferation of ECV304 cells, while methylglyoxal (MG), a toxic metabolite generated during glycolysis and a substrate for Glo I, failed to inhibit the cell growth even at 100 microM. Glo I in several mammalian cell lines was inactivated by GSNO with a pI shift. Although we failed to detect accumulation of MG under conditions of Glo I inactivation, these results suggest that the inhibitory effects of GSNO on cell proliferation and DNA synthesis might be at least partly due to inactivation of Glo I.
Collapse
Affiliation(s)
- A Mitsumoto
- School of Pharmaceutical Sciences, Kitasato University, 5-9-1 Shirokane, Tokyo, 108-8641, Minato-ku, Japan
| | | | | | | |
Collapse
|
143
|
Abstract
Nitric oxide (NO) is a multi-faceted molecule with dichotomous regulatory roles in many areas of biology. The complexity of its biological effects is a consequence of its numerous potential interactions with other molecules such as reactive oxygen species (ROS), metal ions, and proteins. The effects of NO are modulated by both direct and indirect interactions that can be dose-dependent and cell-type specific. For example, in some cell types NO can promote apoptosis, whereas in other cells NO inhibits apoptosis. In hepatocytes, NO can inhibit the main mediators of cell death-caspase proteases. Moreover, low physiological concentrations of NO can inhibit apoptosis, but higher concentrations of NO may be toxic. High NO concentrations lead to the formation of toxic reaction products like dinitrogen trioxide or peroxynitrite that induce cell death, if not by apoptosis, then by necrosis. Long-term exposure to nitric oxide in certain conditions like chronic inflammatory states may predispose cells to tumorigenesis through DNA damage, inhibition of DNA repair, alteration in programmed cell death, or activation of proliferative signaling pathways. Understanding the regulatory mechanisms of NO in apoptosis and carcinogenesis will provide important clues to the diagnosis and treatment of tissue damage and cancer. In this article we have reviewed recent discoveries in the regulatory role of NO in specific cell types, mechanisms of pro-apoptotic and anti-apoptotic induction by NO, and insights into the effects of NO on tumor biology.
Collapse
Affiliation(s)
- P K Kim
- Department of Surgery Laboratories, University of Pittsburgh School of Medicine, PA 15213, USA.
| | | | | | | |
Collapse
|
144
|
Shochina M, Fellig Y, Sughayer M, Pizov G, Vitner K, Podeh D, Hochberg A, Ariel I. Nitric oxide synthase immunoreactivity in human bladder carcinoma. Mol Pathol 2001; 54:248-52. [PMID: 11477140 PMCID: PMC1187076 DOI: 10.1136/mp.54.4.248] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
AIMS To study the expression of the endothelial and inducible isoforms of nitric oxide synthase (eNOS and iNOS, respectively) in human bladder carcinoma and schistosomal bladder disease, and to compare it with normal adult and fetal urothelium. Nitric oxide is thought to play a complex role in human carcinogenesis, but has only recently been investigated in bladder cancer. METHODS Immunohistochemistry was performed on paraffin wax embedded sections of 33 human bladder carcinomas and five bladder carcinoma cell lines; in addition, seven schistosomal bladder cases and normal and fetal urothelium were investigated. In the cell lines enzymatic activity was examined by the NADPH diaphorase reaction. RESULTS Immunoreactivity for eNOS was present in most cells of all 31 cases examined. Immunoreactivity for iNOS was less abundant and was seen in 23 of 25 cases. Similar findings were noted in schistosomal bladder cancer. In the normal bladder mucosa, eNOS immunoreactivity was found only in the superficial cell layer and iNOS was not expressed, whereas in the fetal urothelium immunoreactivity for both isoforms was seen in all cell layers. Enzymatic activity and immunoreactivity for eNOS and iNOS were evident in the five bladder carcinoma cell lines. CONCLUSIONS It is possible that NOS plays a role in the differentiation of the transitional epithelium in fetal life, has a biological function in the adult bladder mucosa, and is involved in bladder carcinogenesis. eNOS and iNOS immunoreactivity do not differ in schistosomal and non-schistosomal bladder carcinoma, but resemble the pattern of expression typical of fetal urothelium.
Collapse
Affiliation(s)
- M Shochina
- Department of Rehabilitation, Hadassah Medical Center and the Hebrew University-Hadassah Medical School, Jerusalem, il-91240, Israel
| | | | | | | | | | | | | | | |
Collapse
|
145
|
Shochina M, Fellig Y, Sughayer M, Pizov G, Vitner K, Podeh D, Hochberg A, Ariel I. Nitric oxide synthase immunoreactivity in human bladder carcinoma. Mol Pathol 2001. [PMID: 11477140 DOI: 10.1136/mp.54.4.248.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS To study the expression of the endothelial and inducible isoforms of nitric oxide synthase (eNOS and iNOS, respectively) in human bladder carcinoma and schistosomal bladder disease, and to compare it with normal adult and fetal urothelium. Nitric oxide is thought to play a complex role in human carcinogenesis, but has only recently been investigated in bladder cancer. METHODS Immunohistochemistry was performed on paraffin wax embedded sections of 33 human bladder carcinomas and five bladder carcinoma cell lines; in addition, seven schistosomal bladder cases and normal and fetal urothelium were investigated. In the cell lines enzymatic activity was examined by the NADPH diaphorase reaction. RESULTS Immunoreactivity for eNOS was present in most cells of all 31 cases examined. Immunoreactivity for iNOS was less abundant and was seen in 23 of 25 cases. Similar findings were noted in schistosomal bladder cancer. In the normal bladder mucosa, eNOS immunoreactivity was found only in the superficial cell layer and iNOS was not expressed, whereas in the fetal urothelium immunoreactivity for both isoforms was seen in all cell layers. Enzymatic activity and immunoreactivity for eNOS and iNOS were evident in the five bladder carcinoma cell lines. CONCLUSIONS It is possible that NOS plays a role in the differentiation of the transitional epithelium in fetal life, has a biological function in the adult bladder mucosa, and is involved in bladder carcinogenesis. eNOS and iNOS immunoreactivity do not differ in schistosomal and non-schistosomal bladder carcinoma, but resemble the pattern of expression typical of fetal urothelium.
Collapse
Affiliation(s)
- M Shochina
- Department of Rehabilitation, Hadassah Medical Center and the Hebrew University-Hadassah Medical School, Jerusalem, il-91240, Israel
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Huang X, Orucevic A, Li M, Gorelik E. Nitric oxide (NO), methylation and TIMP-1 expression in BL6 melanoma cells transfected with MHC class I genes. Clin Exp Metastasis 2001; 18:329-35. [PMID: 11448064 DOI: 10.1023/a:1010867618014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We have previously found that transfection of BL6-8 melanoma cells with the H-2K, but not H-2D/L genes resulted in loss of their metastatic ability that was associated with decrease in their invasiveness and up-regulation of TIMP-1 expression. In the present study using the methylation-specific PCR (MSP) we found that lack of TIMP-1 expression in BL6-8 is associated with methylation in the TIMP-1 5' regulatory area. In the H-2Kb transfected CL8-1 melanoma cells up-regulation of TIMP-1 was in parallel with loss of TIMP-1 gene methylation. Treatment of BL6-8 with 5-azacytidine or with an inhibitor of histone deacetylase trichostatin A resulted in up-regulation of TIMP-1 expression. These results indicate that methylation and histone deacetylation play an important role in transcription repression of TIMP-1 in BL6 melanoma cells. Some data showed that nitric oxide (NO) could affect methylation and expression of various gene. Therefore we analyzed NO production in B16 melanoma cell lines with different expression of TIMP-1. We have found that B16F10 and BL6-8 melanoma cells do not express TIMP-1 and do not produce nitric oxide (NO) even after stimulation with IFN-gamma and LPS. However, BL6-8 cells transfected with H-2Kb or H-2Kd, but not H-2Dd or H-2Ld gene expressed TIMP-1 and produced NO constitutevely. NO production in these cells was further stimulated by IFN-gamma and LPS. Northern blot analysis showed that expression of iNOS was paralleled with TIMP-1 expression in the tested melanoma cells. However, NO produced by SNAP or inhibition of NO production by NMA did not affect TIMP-1 expression in the tested melanoma cells. Thus, TIMP-1 expression and NO production in BL6 melanoma cells transfected with MHC class I gene coincides but it remains unclear whether NO is responsible for the change in TIMP-1 methylation and expression.
Collapse
Affiliation(s)
- X Huang
- University of Pittsburgh Cancer Institute and Department of Pathology, University of Pittsburgh, Pennsylvania 15213, USA
| | | | | | | |
Collapse
|
147
|
Xiong Q, Shi Q, Le X, Wang B, Xie K. Regulation of interleukin-8 expression by nitric oxide in human pancreatic adenocarcinoma. J Interferon Cytokine Res 2001; 21:529-37. [PMID: 11506748 DOI: 10.1089/10799900152434411] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The regulation of interleukin-8 (IL-8) expression by nitric oxide (NO) was determined in human pancreatic cancer cell lines. CaPan-2 and FG human pancreatic adenocarcinoma cells were incubated for 24 h in medium alone or medium containing a cytokine mixture in the presence or absence of an NO synthase (NOS) inhibitor, N(G)-monomethyl-L-arginine (NMA). The NOS activity and level of IL-8 expression were determined. IL-8 expression was induced in the two cell lines. A low level of NOS activity was detectable only in CaPan-2 cells. Moreover, the presence of NMA did not reverse the induction of IL-8. The FG cells were then engineered to produce a physiologic level of NO and incubated in medium alone or medium containing 1 mM NMA. No significant IL-8 expression was induced in those producing a low level of NO, whereas IL-8 expression was induced in those producing a high level of NO. Inhibition of NO production by NMA reversed this effect. Incubation of FG cells with an NO donor, S-nitroso-D,L.-acetyl-penicillamine (SNAP), led to a concentration-dependent and time-dependent induction of IL-8 expression. This NO-mediated upregulation of IL-8 expression correlated with an increase in IL-8 gene transcription and mRNA stability. Our results indicate that NO is involved in the regulation of IL-8 expression in and contributes to the progression of human pancreatic cancer.
Collapse
Affiliation(s)
- Q Xiong
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
148
|
Phillips PG, Birnby LM, Narendran A, Milonovich WL. Nitric oxide modulates capillary formation at the endothelial cell-tumor cell interface. Am J Physiol Lung Cell Mol Physiol 2001; 281:L278-90. [PMID: 11404271 DOI: 10.1152/ajplung.2001.281.1.l278] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide synthase expression has been documented in lung tumors, but a potential role for nitric oxide (NO) in induction of capillary formation remains to be elucidated. The purpose of this report was to characterize the direct effects of NO at the level of the tumor-endothelium interface with respect to angiogenesis. A Transwell two-compartment culture system, human endothelial cells (EC), and two human non-small cell lung cancer (CA) lines that constitutively produce NO were used to simulate the EC-tumor cell interface. Both histological types of lung CA, squamous and adenocarcinoma, induced baseline capillary formation by EC within 3 days. This process was inhibited by NO in the microenvironment because decreasing NO production with 100 microM aminoguanidine (AG) significantly increased capillary formation, whereas coincubation with 100 microM AG plus 400 microM L-arginine returned angiogenesis to baseline values. We demonstrate further that NO may exert its inhibitory effects by influencing matrix metalloproteinase expression/activity and tyrosine phosphorylation of proteins in the sprouting tips of nascent capillaries.
Collapse
Affiliation(s)
- P G Phillips
- Research Service, Samuel S. Stratton Veterans Affairs Medical Center, Albany Medical College, Albany, New York 12208, USA.
| | | | | | | |
Collapse
|
149
|
Massi D, Franchi A, Sardi I, Magnelli L, Paglierani M, Borgognoni L, Maria Reali U, Santucci M. Inducible nitric oxide synthase expression in benign and malignant cutaneous melanocytic lesions. J Pathol 2001; 194:194-200. [PMID: 11400148 DOI: 10.1002/1096-9896(200106)194:2<194::aid-path851>3.0.co;2-s] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide (NO) is synthesized by nitric oxide synthases (NOS) and plays an important role in tumour growth. In this study, inducible NOS (iNOS) expression was evaluated by immunohistochemistry in 34 melanocytic naevi (13 common melanocytic naevi, six Spitz naevi, and 15 so-called 'dysplastic naevi'), ten cutaneous melanomas in situ, 50 stage I invasive melanomas, and eight subcutaneous metastases of melanoma. In addition, four samples of melanocytic naevi and four samples of invasive melanomas were collected in order to perform western blot and northern blot analysis. By immunohistochemistry, melanocytic naevi never expressed iNOS. Among cases of melanoma in situ, two were negative, seven displayed staining in less than 20% of melanoma cells, and positivity was observed in 21-50% of melanoma cells in only one case. iNOS expression was detected in 46 out of 50 invasive melanomas (92%). Among these cases, 18 showed positivity in less than 20% of melanoma cells, 18 showed positivity in 21-50% of melanoma cells, and ten showed iNOS expression in more than 50% of cells. Statistical analysis revealed a significant difference in iNOS expression between melanocytic naevi and cutaneous melanomas (p<0.001). In addition, iNOS expression was significantly higher in invasive melanomas than in melanomas in situ (p=0.01). Among primary cutaneous melanomas, no significant correlation was found between iNOS expression and histopathological parameters (histotype, level, thickness and presence of regression/inflammatory infiltrate) and disease-specific survival. In subcutaneous melanoma metastases, iNOS expression was diffuse in more than 50% of cells. Statistical analysis revealed that subcutaneous melanoma metastases showed greater iNOS immunoreactivity than invasive melanomas (p=0.02). Molecular analyses confirmed that iNOS mRNA and protein were highly expressed in melanoma samples. In conclusion, iNOS was constantly absent in melanocytic naevi, whereas it was frequently expressed in melanomas, with up-regulation of the enzyme paralleling tumour progression. These data suggest that iNOS may play a role in the malignant transformation of melanocytes and in tumour growth. In addition, iNOS may be useful as an immunohistochemical marker for malignant melanocytic lesions.
Collapse
Affiliation(s)
- D Massi
- Dipartimento di Patologia Umana ed Oncologia, Università degli Studi di Firenze, Firenze, Italy
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Choi CY, Kim JY, Kim YS, Chung YC, Seo JK, Jeong HG. Aqueous extract isolated from Platycodon grandiflorum elicits the release of nitric oxide and tumor necrosis factor-alpha from murine macrophages. Int Immunopharmacol 2001; 1:1141-51. [PMID: 11407308 DOI: 10.1016/s1567-5769(01)00047-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Herbal medicines are increasingly being utilized to treat a wide variety of disease processes. Aqueous extract from the root of Platycodon grandiflorum A. DC (Campanulaceae), Changkil (CK), is reported to have antitumor and immunomodulatory activities; however, the mechanism underlying its therapeutic effect is not known. In the present study we examined the effects of CK on the release of nitric oxide (NO) and tumor necrosis factor-alpha (TNF-alpha), and on the gene expression of iNOS and TNF-alpha in mouse macrophages. CK elicited a dose-dependent increase in NO and TNF-alpha production in cultured macrophages. CK significantly affected secretion at concentrations of more than 5 micrograms/ml, and its maximum effect was at concentration of 100 micrograms/ml. Reverse transcription polymerase chain reaction showed that increases in NO and TNF-alpha secretion were due to an increase in inducible NO synthase mRNA and TNF-alpha mRNA, respectively. Transient expression assays with NF-kappa B binding sites linked to the luciferase gene revealed that CK-induced increase of inducible NO synthase mRNA and TNF-alpha mRNA were mediated by the NF-kappa B transcription factor complex. These results demonstrate that CK stimulates NO and TNF-alpha release and is able to upregulate iNOS and TNF-alpha expression through NF-kappa B transactivation and this may be a mechanism whereby this herbal medicine elicits its therapeutic effects.
Collapse
Affiliation(s)
- C Y Choi
- Department of Pharmacy and Research Center for Proteineous Materials, Chosun University, 375 Seosuk-dong, Gong-ku, Kwangju 501-759, South Korea
| | | | | | | | | | | |
Collapse
|