101
|
Cherng JM, Tsai KD, Perng DS, Wang JS, Wei CC, Lin JC. Diallyl sulfide protects against ultraviolet B-induced skin cancers in SKH-1 hairless mouse: analysis of early molecular events in carcinogenesis. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2011; 27:138-46. [DOI: 10.1111/j.1600-0781.2011.00582.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
102
|
Bhattacharya A. Methylselenocysteine: a promising antiangiogenic agent for overcoming drug delivery barriers in solid malignancies for therapeutic synergy with anticancer drugs. Expert Opin Drug Deliv 2011; 8:749-63. [PMID: 21473705 DOI: 10.1517/17425247.2011.571672] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Despite progress, chemotherapeutic response in solid malignancies has remained limited. Although initial results of the use of antiangiogenic agents in combination chemotherapy indicated an enhanced therapeutic response, recent data indicate that the surviving cancer is not only able to surmount therapy, but also actually able to adapt a more aggressive metastatic phenotype. Thus, selecting an antiangiogenic agent that is less likely to lead to tumor resurgence is a key to future therapeutic success of antiangiogenic agents in a combinatorial setting. AREAS COVERED Against the broad spectrum of antiangiogenic agents used at present in the clinic, the putative benefits of the use of organoselenium compounds, such as methylselenocysteine (MSC), are discussed in this review. EXPERT OPINION MSC, being part of the mammalian physiology, is a well-tolerated, versatile and economical antiangiogenic agent. It downregulates multiple key upstream tumor survival markers, and enhances tumor drug delivery, at a given systemic dose of an anticancer agent, while protecting normal tissue from cytotoxic adverse effects. Further clinical trials, especially in poorly differentiated cancers, are warranted.
Collapse
Affiliation(s)
- Arup Bhattacharya
- Roswell Park Cancer Institute, Department of Cancer Prevention and Control, Buffalo, NY 14263, USA.
| |
Collapse
|
103
|
Zhang W, He XJ, Ma YY, Wang HJ, Xia YJ, Zhao ZS, Ye ZY, Tao HQ. Inducible nitric oxide synthase expression correlates with angiogenesis, lymphangiogenesis, and poor prognosis in gastric cancer patients. Hum Pathol 2011; 42:1275-82. [PMID: 21333324 DOI: 10.1016/j.humpath.2010.09.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 08/27/2010] [Accepted: 09/02/2010] [Indexed: 01/28/2023]
Abstract
Increased nitric oxide synthase expression plays a key role in tumor progression. To examine inducible nitric oxide synthase expression and its correlation with clinical variables, such as tumor progression, angiogenesis, lymphangiogenesis, and prognosis in gastric cancer, we studied inducible nitric oxide synthase expression in gastric cancer samples from 211 patients with 5-year follow-up. CD105 and D2-40 were adopted as biomarkers for tumor angiogenesis and lymphangiogenesis, respectively. Inducible nitric oxide synthase staining was mainly found in the cytoplasm of gastric cancer tumor cells. Positive inducible nitric oxide synthase immunoreactivity was seen in 54.03% of gastric cancer specimens, which was correlated with lymph node metastasis, vascular invasion, distant metastasis, and TNM stage. Compared with inducible nitric oxide synthase negative patients, inducible nitric oxide synthase-positive patients had significantly shorter survival times and higher microvessel density and lymphatic vessel density. Intratumor and peritumor blood microvessel density and lymphatic vessel density correlated with inducible nitric oxide synthase expression (Spearman ρ test, P < .05). We conclude that inducible nitric oxide synthase expression correlates with lymph node metastasis, vascular invasion, distant metastasis, TNM stage, and poor survival rate in gastric cancer. We propose that synthesized inducible nitric oxide synthase increases angiogenesis, and lymphangiogenesis thus promotes tumor progression. Inducible nitric oxide synthase expression may be a good biomarker for poor prognosis in gastric cancer.
Collapse
Affiliation(s)
- Wei Zhang
- Wenzhou Medical College, Wenzhou, 325035, China
| | | | | | | | | | | | | | | |
Collapse
|
104
|
Roy S, Reddy BS, Sudhakar G, Kumar JM, Banerjee R. 17β-estradiol-linked nitro-L-arginine as simultaneous inducer of apoptosis in melanoma and tumor-angiogenic vascular endothelial cells. Mol Pharm 2011; 8:350-9. [PMID: 21291219 DOI: 10.1021/mp2000346] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aggressive melanoma is commonly associated with rapid angiogenic growth in tumor mass, tumor cells acquiring apoptosis resistance, inhibition of cellular differentiation etc. Designing a single anticancer molecule which will target all these factors simultaneously is challenging. In the pretext of inciting anticancer effect through inhibiting nitric oxide synthase (NOS) via estrogen receptors (ER) in ER-expressing skin cancer cells, we developed an estrogen-linked L-nitro-arginine molecule (ESAr) for inciting anticancer effect in melanoma cells. ESAr showed specific anticancer effect through diminishing aggressiveness and metastatic behavior in melanoma cells and tumor. In comparison, ESAr showed significantly higher antiproliferative effect than parent molecule L-nitroarginine methyl ester (L-NAME, a NOS inhibitor) through induction of prominent apoptosis in melanoma cells. ESAr-pretreated aggressive melanoma cells could not form tumor possibly because of transformation/differentiation into epithelial-type cells. Furthermore, its antiangiogenic effect was demonstrated through ESAr-induced antiproliferation in HUVEC cells and apoptosis-induction in tumor-associated vascular endothelial cells, thereby significantly restricting severe growth in melanoma tumor. The targeting moiety, estrogen, at the therapeutic concentration of ESAr has apparently no effect in tumor-growth reduction. Albeit, no specific NOS-inhibition was observed, but ESAr could simultaneously induce these three cancer-specific antiaggressiveness factors, which the parent molecule could not induce. Our data rationalize and establish a new use of estrogen as a ligand for potentially targeting multiple cellular factors for treating aggressive cancers.
Collapse
Affiliation(s)
- Sayantani Roy
- Division of Lipid Science & Technology, Indian Institute of Chemical Technology, Hyderabad, Andhra Pradesh 500007, India
| | | | | | | | | |
Collapse
|
105
|
Arif M, Vedamurthy BM, Choudhari R, Ostwal YB, Mantelingu K, Kodaganur GS, Kundu TK. Nitric oxide-mediated histone hyperacetylation in oral cancer: target for a water-soluble HAT inhibitor, CTK7A. ACTA ACUST UNITED AC 2011; 17:903-13. [PMID: 20797619 DOI: 10.1016/j.chembiol.2010.06.014] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 05/27/2010] [Accepted: 06/03/2010] [Indexed: 12/30/2022]
Abstract
Altered histone acetylation is associated with several diseases, including cancer. We report here that, unlike in most cancers, histones are found to be highly hyperacetylated in oral squamous cell carcinoma (OSCC; oral cancer) patient samples. Mechanistically, overexpression, as well as enhanced autoacetylation, of p300 induced by nucleophosmin (NPM1) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) causes the hyperacetylation, which is nitric oxide (NO) signal dependent. Inhibition of the histone acetyltransferase (HAT) activity of p300 by a water-soluble, small molecule inhibitor, Hydrazinocurcumin (CTK7A), substantially reduced the xenografted oral tumor growth in mice. These results, therefore, not only establish an epigenetic target for oral cancer, but also implicate a HAT inhibitor (HATi) as a potential therapeutic molecule.
Collapse
Affiliation(s)
- Mohammed Arif
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Jakkur PO, Bangalore-560 064, Karnataka, India
| | | | | | | | | | | | | |
Collapse
|
106
|
Guruvayoorappan C, Kuttan G. Inhibition of tumor specific angiogenesis by amentoflavone. BIOCHEMISTRY (MOSCOW) 2011; 73:209-18. [DOI: 10.1134/s0006297908020132] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
107
|
Muntané J, la Mata MD. Nitric oxide and cancer. World J Hepatol 2010; 2:337-44. [PMID: 21161018 PMCID: PMC2999298 DOI: 10.4254/wjh.v2.i9.337] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Revised: 09/02/2010] [Accepted: 09/09/2010] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) is a lipophilic, highly diffusible and short-lived physiological messenger which regulates a variety of important physiological responses including vasodilation, respiration, cell migration, immune response and apoptosis. NO is synthesized by three differentially gene-encoded NO synthase (NOS) in mammals: neuronal NOS (nNOS or NOS-1), inducible NOS (iNOS or NOS-2) and endothelial NOS (eNOS or NOS-3). All isoforms of NOS catalyze the reaction of L-arginine, NADPH and oxygen to NO, L-citrulline and NADP. NO may exert its cellular action by cGMP-dependent as well as by cGMP-independent pathways including postranslational modifications in cysteine (S-nitrosylation or S-nitrosation) and tyrosine (nitration) residues, mixed disulfide formation (S-nitrosoglutathione or GSNO) or promoting further oxidation protein stages which have been related to altered protein function and gene transcription, genotoxic lesions, alteration of cell-cycle check points, apoptosis and DNA repair. NO sensitizes tumor cells to chemotherapeutic compounds. The expression of NOS-2 and NOS-3 has been found to be increased in a variety of human cancers. The multiple actions of NO in the tumor environment is related to heterogeneous cell responses with particular attention in the regulation of the stress response mediated by the hypoxia inducible factor-1 and p53 generally leading to growth arrest, apoptosis or adaptation.
Collapse
Affiliation(s)
- Jordi Muntané
- Jordi Muntané, Liver Research Unit, Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), "Reina Sofia" University Hospital, Cordoba E-14004, Spain
| | | |
Collapse
|
108
|
Martinez-Outschoorn UE, Balliet RM, Rivadeneira DB, Chiavarina B, Pavlides S, Wang C, Whitaker-Menezes D, Daumer KM, Lin Z, Witkiewicz AK, Flomenberg N, Howell A, Pestell RG, Knudsen ES, Sotgia F, Lisanti MP. Oxidative stress in cancer associated fibroblasts drives tumor-stroma co-evolution: A new paradigm for understanding tumor metabolism, the field effect and genomic instability in cancer cells. Cell Cycle 2010; 9:3256-76. [PMID: 20814239 DOI: 10.4161/cc.9.16.12553] [Citation(s) in RCA: 352] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Loss of stromal fibroblast caveolin-1 (Cav-1) is a powerful single independent predictor of poor prognosis in human breast cancer patients, and is associated with early tumor recurrence, lymph node metastasis and tamoxifen-resistance. We developed a novel co-culture system to understand the mechanism(s) by which a loss of stromal fibroblast Cav-1 induces a "lethal tumor micro-environment." Here, we propose a new paradigm to explain the powerful prognostic value of stromal Cav-1. In this model, cancer cells induce oxidative stress in cancer-associated fibroblasts, which then acts as a "metabolic" and "mutagenic" motor to drive tumor-stroma co-evolution, DNA damage and aneuploidy in cancer cells. More specifically, we show that an acute loss of Cav-1 expression leads to mitochondrial dysfunction, oxidative stress and aerobic glycolysis in cancer associated fibroblasts. Also, we propose that defective mitochondria are removed from cancer-associated fibroblasts by autophagy/mitophagy that is induced by oxidative stress. As a consequence, cancer associated fibroblasts provide nutrients (such as lactate) to stimulate mitochondrial biogenesis and oxidative metabolism in adjacent cancer cells (the "Reverse Warburg Effect"). We provide evidence that oxidative stress in cancer-associated fibroblasts is sufficient to induce genomic instability in adjacent cancer cells, via a bystander effect, potentially increasing their aggressive behavior. Finally, we directly demonstrate that nitric oxide (NO) over-production, secondary to Cav-1 loss, is the root cause for mitochondrial dysfunction in cancer associated fibroblasts. In support of this notion, treatment with anti-oxidants (such as N-acetyl-cysteine, metformin and quercetin) or NO inhibitors (L-NAME) was sufficient to reverse many of the cancer-associated fibroblast phenotypes that we describe. Thus, cancer cells use "oxidative stress" in adjacent fibroblasts (i) as an "engine" to fuel their own survival via the stromal production of nutrients and (ii) to drive their own mutagenic evolution towards a more aggressive phenotype, by promoting genomic instability. We also present evidence that the "field effect" in cancer biology could also be related to the stromal production of ROS and NO species. eNOS-expressing fibroblasts have the ability to downregulate Cav-1 and induce mitochondrial dysfunction in adjacent fibroblasts that do not express eNOS. As such, the effects of stromal oxidative stress can be laterally propagated, amplified and are effectively "contagious"--spread from cell-to-cell like a virus--creating an "oncogenic/mutagenic" field promoting widespread DNA damage.
Collapse
|
109
|
Cao T, Zhang T, Wang L, Zhang L, Adachi T, Sato T, Yamagata S, Yamagata T. Ganglioside GD1a Suppression of NOS2 Expression Via ERK1 Pathway in Mouse Osteosarcoma FBJ Cells. J Cell Biochem 2010; 110:1165-74. [DOI: 10.1002/jcb.22631] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
110
|
Cheenpracha S, Park EJ, Yoshida WY, Barit C, Wall M, Pezzuto JM, Chang LC. Potential anti-inflammatory phenolic glycosides from the medicinal plant Moringa oleifera fruits. Bioorg Med Chem 2010; 18:6598-602. [PMID: 20685125 DOI: 10.1016/j.bmc.2010.03.057] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2010] [Accepted: 03/23/2010] [Indexed: 11/17/2022]
Abstract
Bioassay-guided isolation and purification of the ethyl acetate extract of Moringa oleifera fruits yielded three new phenolic glycosides; 4-[(2'-O-acetyl-alpha-l-rhamnosyloxy) benzyl]isothiocyanate (1), 4-[(3'-O-acetyl-alpha-l-rhamnosyloxy)benzyl]isothiocyanate (2), and S-methyl-N-{4-[(alpha-l-rhamnosyloxy)benzyl]}thiocarbamate (3), together with five known phenolic glycosides (4-8). The structures of the new metabolites were determined on the basis of spectroscopic analyses including 1D- and 2D-NMR and mass spectrometry. The anti-inflammatory activity of isolated compounds was investigated with the lipopolysaccharide (LPS)-induced murine macrophage RAW 264.7 cell line. It was found that 4-[(2'-O-acetyl-alpha-l-rhamnosyloxy)benzyl]isothiocyanate (1) possessed potent NO-inhibitory activity with an IC(50) value of 1.67 microM, followed by 2 (IC(50)=2.66 microM), 4 (IC(50)=2.71 microM), and 5 (IC(50)=14.4 microM), respectively. Western blots demonstrated these compounds reduced LPS-mediated iNOS expression. In the concentration range of the IC(50) values, no significant cytotoxicity was noted. Structure-activity relationships following NO-release indicated: (1) the isothiocyanate group was essential for activity, (2) acetylation of the isothiocyanate derivatives at C-2' or at C-3' of rhamnose led to higher activity, (3) un-acetylated isothiocyanate derivatives displayed eight times less activity than the acetylated derivatives, and (4) acetylation of the thiocarbamate derivatives enhanced activity. These data indicate compounds 1, 2, 4 and 5 are responsible for the reported NO-inhibitory effect of Moringa oleifera fruits, and further studies are warranted.
Collapse
Affiliation(s)
- Sarot Cheenpracha
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawaii at Hilo, 34 Rainbow Drive, Hilo, HI 96720, USA
| | | | | | | | | | | | | |
Collapse
|
111
|
Huang YJ, Zhang BB, Ma N, Murata M, Tang AZ, Huang GW. Nitrative and oxidative DNA damage as potential survival biomarkers for nasopharyngeal carcinoma. Med Oncol 2010; 28:377-84. [PMID: 20339958 DOI: 10.1007/s12032-010-9434-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 01/25/2010] [Indexed: 12/14/2022]
Abstract
Currently, there are no satisfactory biomarkers available to screen for nasopharyngeal carcinoma (NPC). Nitric oxide (NO), produced by inducible nitric oxide synthase (iNOS), has been suggested to cause nitrative and oxidative stress, leading to the accumulation of 8-nitroguanine (8-NitroG) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) and the subsequent transversion mutation of DNA. The aim of this study was to evaluate iNOS expression and the status of nitrative and oxidative stress in NPC. Fifty-nine cases of NPC and 39 cases of chronic nasopharyngitis were investigated to examine the expression of iNOS and the formation of 8-NitroG and 8-OHdG, using double-immunofluorescent staining. The statistical differences in immunoreactivities were analyzed using the Mann-Whitney test. Thirty-six patients from the 57 cases of NPC and 36 healthy controls were investigated to examine the level of serum 8-OHdG, using enzyme-linked immunosorbent assay (ELISA). The statistical differences were analyzed using a t test. Strong DNA lesions were observed in the cancer cells of NPC patients. All cases of NPC were positive for 8-NitroG and 8-OHdG, and 54 (94.7%) were positive for iNOS. NPC samples exhibited significantly more intense staining for 8-NitroG, 8-OHdG and iNOS than those of chronic nasopharyngitis (P < 0.05, respectively). The mean value of serum 8-OHdG in the 36 NPC patients was 0.538 ± 0.336 ng/ml compared to 0.069 ± 0.059 ng/ml for the healthy controls. The difference in the serum levels of 8-OHdG between the NPC patients and controls was statistically significant (P < 0.05). Our present findings suggest that pathological stimulation of nasopharyngeal tissue, caused by bacterial, viral or parasitic inflammation, may lead to nitrative and oxidative DNA lesions, caused by NO. This may contribute to the cause and development of NPC. Thus, 8-NitroG and 8-OHdG could be potential biomarkers for evaluating the risk of NPC. Better understanding of the molecular mechanisms underlying nitrative and oxidative DNA damage may provide clues to molecular targets for new approaches of NPC prevention.
Collapse
Affiliation(s)
- Yuan-Jiao Huang
- Medical Scientific Research Center, Guangxi Medical University, 530021 Nanning, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
112
|
Zafirellis K, Zachaki A, Agrogiannis G, Gravani K. Inducible nitric oxide synthase expression and its prognostic significance in colorectal cancer. APMIS 2010; 118:115-24. [PMID: 20132175 DOI: 10.1111/j.1600-0463.2009.02569.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nitric oxide synthases (NOS) are expressed in colorectal cancer. The aim of this study was to examine the inducible NOS (iNOS) expression in colorectal cancer and to investigate its prognostic relevance. Tissue sections of primary tumors from 132 patients undergoing curative resection for colorectal cancer were immunohistochemically examined for iNOS expression. The expression pattern of iNOS was correlated with various clinicopathological characteristics and survival. iNOS immunoreactivity was observed in the cytoplasm of tumor epithelial cells in 60 patients (45.5%) and positively correlated with lymph node involvement (p = 0.019). No significant correlation was found between iNOS expression and various clinicopathological characteristics, including age, gender, tumor location, tumor size, tumor grade, T stage, and Union International Contra la Cancrum (UICC) stage. Survival analysis showed a significant correlation between iNOS-positive tumors and poor disease-specific survival (p < 0.0001), with independent prognostic significance in multivariate analysis (HR = 4.42; p < 0.0001). Patients with stage II disease and iNOS-positive tumors had significantly worse disease-specific survival than those with iNOS-negative tumors (p < 0.0001). In addition, patients with stage III disease and iNOS-positive tumors had significantly worse disease-specific survival than those with iNOS-negative tumors (p = 0.001). The ability of iNOS to predict outcome in colorectal cancer patients may be independent of other known prognostic factors, providing a new molecular marker with significant potential for clinical utility.
Collapse
|
113
|
Tiwari SK, Manoj G, Sharma V, Sivaram G, Saikant R, Bardia A, Sharma VK, Abid Z, Khan AA, Habeeb MA, Habibullah CM, Kumar BS, Nandan A. Relevance of Helicobacter pylori genotypes in gastric pathology and its association with plasma malondialdehyde and nitric oxide levels. Inflammopharmacology 2010; 18:59-64. [PMID: 20143166 DOI: 10.1007/s10787-010-0031-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 01/11/2010] [Indexed: 12/12/2022]
Abstract
Persistent infection with Helicobacter pylori confers an increased risk of peptic ulceration and gastric adenocarcinoma. Reactive oxygen and nitrogen species play a crucial role in the progression from normal gastric mucosa to cancer. The aim of the present study was to investigate the plasma malondialdehyde and nitric oxide levels in H. pylori related gastroduodenal diseases and associate their levels with gastric pathology and genotypes of H. pylori. Malondialdehyde and nitric oxide levels in plasma samples of 250 subjects were spectrophotometrically determined. Subsequently, genotypic and histopathological assessment was performed in gastric biopsies obtained during endoscopy. The levels of MDA and NO exceeded in subjects infected with genotype-1 of Hp than those with other genotypes suggesting more precise interaction of highly virulent strains of Hp in eliciting severe tissue damage. In conclusion, the study demonstrates close relationship between the plasma malondialdehyde and nitric oxide levels, gastric histopathology and genotypes of H. pylori.
Collapse
Affiliation(s)
- Santosh K Tiwari
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058, Andhra Pradesh, India
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Prabhu V, Guruvayoorappan C. Nitric oxide: pros and cons in tumor progression. Immunopharmacol Immunotoxicol 2010; 32:387-92. [DOI: 10.3109/08923970903440192] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
115
|
Yoon T, Cheon MS, Lee AY, Lee DY, Moon BC, Chun JM, Choo BK, Kim HK. Anti-inflammatory Activity of Methylene Chloride Fraction From Glehnia littoralis Extract via Suppression of NF-κB and Mitogen-Activated Protein Kinase Activity. J Pharmacol Sci 2010; 112:46-55. [DOI: 10.1254/jphs.09168fp] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
116
|
Bui-Nguyen TM, Pakala SB, Sirigiri DR, Martin E, Murad F, Kumar R. Stimulation of inducible nitric oxide by hepatitis B virus transactivator protein HBx requires MTA1 coregulator. J Biol Chem 2009; 285:6980-6. [PMID: 20022949 DOI: 10.1074/jbc.m109.065987] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nitric oxide has been implicated in the pathogenesis of inflammatory disorders, including hepatitis B virus-associated hepatocellular carcinoma. Transactivator protein HBx, a major regulator of cellular responses of hepatitis B virus, is known to induce the expression of MTA1 (metastasis-associated protein 1) coregulator via NF-kappaB signaling in hepatic cells. However, the underlying mechanism of HBx regulation of the inducible nitric-oxide synthase (iNOS) pathway remains unknown. Here we provide evidence that MTA1 is a positive regulator of iNOS transcription and plays a mechanistic role in HBx stimulation of iNOS expression and activity. We found that the HBx-MTA1 complex is recruited onto the human iNOS promoter in an NF-kappaB-dependent manner. Pharmacological inhibition of the NF-kappaB signaling prevented the ability of HBx to stimulate the transcription, the expression, and the activity of iNOS; nevertheless, these effects could be substantially rescued by MTA1 dysregulation. We further discovered that HBx-mediated stimulation of MTA1 is paralleled by the suppression of miR-661, a member of the small noncoding RNAs, recently shown to target MTA1. We observed that miR-661 controls of MTA1 expression contributed to the expression and activity of iNOS in HBx-expressing HepG2 cells. Accordingly, depletion of MTA1 by either miR-661 or siRNA in HBx-expressing cells severely impaired the ability of HBx to modulate the endogenous levels of iNOS and nitrite production. Together, these findings reveal an inherent role of MTA1 in HBx regulation of iNOS expression and consequently its function in the liver cancer cells.
Collapse
Affiliation(s)
- Tri M Bui-Nguyen
- Department of Biochemistry and Molecular Biology, George Washington University Medical Center, Washington, DC 20037, USA
| | | | | | | | | | | |
Collapse
|
117
|
Wang S, Zhang Z, Lin X, Xu DS, Feng Y, Ding K. A polysaccharide, MDG-1, induces S1P1 and bFGF expression and augments survival and angiogenesis in the ischemic heart. Glycobiology 2009; 20:473-84. [PMID: 20008963 DOI: 10.1093/glycob/cwp199] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Ophiopogon japonicus is a traditional Chinese medicine used to treat cardiovascular disease. Recent studies have confirmed its beneficial properties, but not the mechanism of action. Herein, we investigate the anti-ischemic properties of a water-soluble beta-d-fructan (MDG-1) from Ophiopogon japonicus, and assess the cytoprotective and proangiogenic effects of MDG-1. MDG-1 protects cardiomyocyte and microvascular endothelial cells (HMEC-1) against oxygen glucose deprivation (OGD)-induced cell death, as well as protect myocardial cells from ischemia-induced death occurring after coronary artery ligation in rats. Meanwhile, MDG-1 stimulates the differentiation of HMEC-1 cells into capillary-like structures in vitro and functions as a chemoattractant in migration assays, and promotes neovascularization in ischemic myocardium. In addition, MDG-1 upregulates sphingosine kinase 1 and sphingosine-1-phosphate (S1P) receptor 1 expression. Both MDG-1 and S1P induce basic fibroblast growth factor (bFGF) expression in HMEC-1 cells. Further study revealed that both MDG-1 and S1P induce Akt and ERK phosphorylation in a dose- and time-dependent manner, an effect that is attenuated by pre-treatment with either the Akt inhibitor wortmannin or the ERK inhibitor PD98059, and MDG-1 can also induce eNOS phosphorylation and increases in production of NO. These data indicate that MDG-1 presented remarkable anti-ischemic activity and protects cardiomyocyte and HMEC-1 cells from ischemia-induced cell damage by inducing S1P1 and bFGF cytoprotective and proangiogenic effects via the S1P/bFGF/Akt/ERK/eNOS signaling pathway.
Collapse
Affiliation(s)
- Shuo Wang
- Engineering Research Center of Modern Preparation Technology of TCM, Shanghai University of Traditional Chinese Medicine, Pudong, Shanghai, 201203, People's Republic of China
| | | | | | | | | | | |
Collapse
|
118
|
Melisi D, Rosso F, Curcio A, Tortora C, Nieddu M, Marino G, Lettieri M, Grimaldi A, Luongo E, Romano S, Romano MF, Boatto G, Abignente E, Barbarisi A, Rimoli MG. Galactosyl derivative ofNω-nitro-L-arginine: Study of antiproliferative activity on human thyroid follicular carcinoma cells. J Cell Physiol 2009; 221:440-7. [DOI: 10.1002/jcp.21876] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
119
|
Pitiyage G, Tilakaratne WM, Tavassoli M, Warnakulasuriya S. Molecular markers in oral epithelial dysplasia: review. J Oral Pathol Med 2009; 38:737-52. [DOI: 10.1111/j.1600-0714.2009.00804.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
120
|
Nematollahi S, Nematbakhsh M, Haghjooyjavanmard S, Khazaei M, Salehi M. Inducible nitric oxide synthase modulates angiogenesis in ischemic hindlimb of rat. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2009; 153:125-9. [PMID: 19771137 DOI: 10.5507/bp.2009.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Angiogenesis plays an important role in maintaining adequate oxygen delivery, and nitric oxide (NO) is a potential regulator of angiogenesis. NO is synthesized through three isoforms of NO synthase (NOS). It is hypothesized that the NO derived from inducible NOS (iNOS) may promote survival of ischemic tissue through angiogenesis. To test this hypothesis, we investigated the effect of iNOS deficiency (by L-NIL) on angiogenesis in a hindlimb ischemia model. METHODS Thirty-two male wistar rats randomly divided into four groups. In groups 1 & 2, hindlimb ischemia was induced by ligation of femoral artery and they received L-NIL and saline respectively. The animals in groups 3 and 4 also received L-NIL and saline respectively without surgical procedure. After 21 days, the serum concentration of nitrite, capillary density and expression of HIF1alpha were determined. RESULTS Serum nitrite levels were significantly lower in L-NIL groups (p<0.05). The capillary density in group 1 (ischemia+L-NIL) was significantly different from group 2 (ischemia+saline); group 1: 360.33+/-77.02, group 2: 549+/-81.85 /mm2, p<0.05) .In addition, expression of HIF1alpha was significantly increased in ischemic groups (p<0.05). CONCLUSION Selective inhibition of iNOS by L-NIL inhibits angiogenesis in a hindlimb ischemic rat model. In addition, ischemia induces expression of HIF1alpha in hypoxic tissue.
Collapse
Affiliation(s)
- Sedigheh Nematollahi
- Department of Physiology, Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | | | | | | |
Collapse
|
121
|
Yan L, Chen WL, Zeng SG, Wang YJ, Bai ZB. Inhibition of VEGF expression in tongue squamous cancer cells via RNA interference silencing of iNOS gene. Int J Oral Maxillofac Surg 2009; 38:369-73. [PMID: 19307105 DOI: 10.1016/j.ijom.2009.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2008] [Revised: 04/16/2008] [Accepted: 01/29/2009] [Indexed: 10/21/2022]
Abstract
The purpose of this study was to investigate the regulatory role of the inducible nitric oxide synthase (iNOS) gene on vascular endothelial growth factor (VEGF) expression in oral squamous cancer cells. The RNA interference (RNAi) technique was used to silence iNOS gene expression by transfecting an expression vector containing short hairpin RNA (shRNA) for iNOS into Tca8113 tongue squamous cancer cells using cationic liposomes. Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blotting were used to monitor iNOS and VEGF mRNA, as well as protein expression. iNOS mRNA expression was significantly downregulated 24 and 36 h after transfection, and iNOS protein expression was significantly downregulated at 36 and 48 h (P<0.05 versus control), showing that effective silencing was achieved. VEGF mRNA was significantly decreased 24 and 36 h post-transfection, and VEGF protein expression was significantly decreased at 36 and 48 h (P<0.05). RNAi can decrease iNOS gene expression and achieve a gene silencing effect. iNOS gene silencing reduces VEGF expression levels in Tca8113 cells. Thus, there is a relationship between iNOS and VEGF expression in tongue squamous cancer cells.
Collapse
Affiliation(s)
- Lan Yan
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou, Guangdong 510120, PR China
| | | | | | | | | |
Collapse
|
122
|
Han X, Zheng T, Lan Q, Zhang Y, Kilfoy BA, Qin Q, Rothman N, Zahm SH, Holford TR, Leaderer B, Zhang Y. Genetic polymorphisms in nitric oxide synthase genes modify the relationship between vegetable and fruit intake and risk of non-Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev 2009; 18:1429-38. [PMID: 19423521 DOI: 10.1158/1055-9965.epi-09-0001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Oxidative damage caused by reactive oxygen species and other free radicals is involved in carcinogenesis. It has been suggested that high vegetable and fruit intake may reduce the risk of non-Hodgkin lymphoma (NHL) as vegetables and fruit are rich in antioxidants. The aim of this study is to evaluate the interaction of vegetable and fruit intake with genetic polymorphisms in oxidative stress pathway genes and NHL risk. This hypothesis was investigated in a population-based case-control study of NHL and NHL histologic subtypes in women from Connecticut, including 513 histologically confirmed incident cases and 591 randomly selected controls. Gene-vegetable/fruit joint effects were estimated using unconditional logistic regression model. The false discovery rate method was applied to adjust for multiple comparisons. Significant interactions with vegetable and fruit intake were mainly found for genetic polymorphisms on nitric oxide synthase (NOS) genes among those with diffuse large B-cell lymphoma and follicular lymphoma. Two single nucleotide polymorphisms in the NOS1 gene were found to significantly modify the association between total vegetable and fruit intake and risk of NHL overall, as well as the risk of follicular lymphoma. When vegetables, bean vegetables, cruciferous vegetables, green leafy vegetables, red vegetables, yellow/orange vegetables, fruit, and citrus fruits were examined separately, strong interaction effects were narrowed to vegetable intake among patients with diffuse large B-cell lymphoma. Our results suggest that genetic polymorphisms in oxidative stress pathway genes, especially in the NOS genes, modify the association between vegetable and fruit intake and risk of NHL.
Collapse
Affiliation(s)
- Xuesong Han
- Yale University School of Public Health, New Haven, CT 06520-8034, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Avci A, Tüzüner-Oncül AM, Gökcan MK, Namuslu M, Oztürk A, Durak I. Nitric oxide metabolism in cancerous and non-cancerous oral gingivomucosal tissues: possible implications of nitric oxide in cancer process. J Oral Pathol Med 2009; 38:304-6. [PMID: 19141063 DOI: 10.1111/j.1600-0714.2008.00719.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Nitric oxide (NO) is a molecule that plays various roles in the body tissues. NO plays important roles in vasodilatation, platelet aggregation, cytokine stimulation, neurotransmission, immune function, etc. NO also exerts dual functions as an oxidant and antioxidant substance depending on its concentrations and environmental conditions. In this study, we aimed to examine possible correlation between NO levels and NO synthase (NOS) activity in the patients with oral cancer. MATERIALS AND METHODS The study included 19 tissues from human subjects (11 malign and eight benign lesions). RESULTS NO level and NOS activity were found decreased in the malign lesions compared with those of the benign ones. CONCLUSIONS In conclusion, two suggestions can be made; first, decreased NO synthesis may be an attempt to suppress angiogenesis, which is known to provide more essential nutrients to malign lesions and/or second, malign lesions may suppress NO production to be capable of creating more rapid proliferation as it has been known that NO is also a powerful free radical inducing apoptosis.
Collapse
Affiliation(s)
- A Avci
- Department of Biochemistry, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | | | | | | | | | | |
Collapse
|
124
|
Chen CN, Lin JJ, Lee H, Cheng YM, Chang KJ, Hsieh FJ, Lai HS, Chang CC, Lee PH. Association between color doppler vascularity index, angiogenesis-related molecules, and clinical outcomes in gastric cancer. J Surg Oncol 2009; 99:402-8. [DOI: 10.1002/jso.21193] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
125
|
Zhang Q, Zhang J, Guan Y, Zhang S, Zhu C, Xu GT, Wang L. Suppression of retinal neovascularization by the iNOS inhibitor aminoguanidine in mice of oxygen-induced retinopathy. Graefes Arch Clin Exp Ophthalmol 2009; 247:919-27. [PMID: 19301028 DOI: 10.1007/s00417-009-1066-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/02/2009] [Accepted: 02/26/2009] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Retinal neovascularization (NV) is a major cause of blindness associated with ischemic retinal disorders. Our study was focused on evaluating the inhibitory effect of aminoguanidine (AG), an inhibitor of inducible nitric oxide synthase (iNOS), on retinal NV in mice of oxygen-induced retinopathy (OIR). METHODS An OIR model was established with 7-day-old C57BL/6J mice. One day before and 1 and 3 days after being returned to the room air, the right eyes were injected intravitreally with bevacizumab, AG or bevacizumab+AG respectively. The left eyes were injected with normal saline (NS) as control. The mice were killed at postnatal day 17 (P17). The effects of AG or bevacizumab on iNOS or VEGF expressions were evaluated by RT-PCR and immunohistochemistry. Retinal NV was examined by fluorescein angiography, and was quantified histologically by CD34 immnunostaining at P17. RESULTS Compared with NS-treated eyes, retinal VEGF and iNOS mRNA expressions were significantly reduced in AG- and bevacizumab+AG-treated eyes; whereas in bevacizumab-treated eyes, retinal VEGF mRNA expression increased and iNOS mRNA expression remained unchanged. The above changes were confirmed by immunohistochemical study. The generalized decrease in both VEGF and iNOS distributions in mice retina treated with AG or bevacizumab+AG was demonstrated by immunohistochemistry. Retinal NV was significantly reduced in all three groups treated with bevacizumab, AG or bevacizumab+AG, when compared with NS-treated eyes. CONCLUSIONS iNOS activation plays a pathological role in retinal NV in a mouse model of ischemic retinopathy. Administration of AG significantly suppressed retinal NV. Therefore, AG appears to be a novel and effective therapeutic approach for retinal NV.
Collapse
Affiliation(s)
- Qiong Zhang
- Department of Ophthalmology, Ruijin Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
126
|
Punathil T, Katiyar SK. Inhibition of non-small cell lung cancer cell migration by grape seed proanthocyanidins is mediated through the inhibition of nitric oxide, guanylate cyclase, and ERK1/2. Mol Carcinog 2009; 48:232-242. [PMID: 18680102 DOI: 10.1002/mc.20473] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tumor cell migration is considered as a major event in the metastatic cascade. Here we examined the effect of grape seed proanthocyanidins (GSPs) on migration capacity and signaling mechanisms using nonsmall cell human lung cancer cells. Using in vitro migration assay, we found that treatment of A549 and H1299 cells with GSPs resulted in concentration-dependent inhibition of migration of these cells. The migration capacity of cells was reduced in presence of N(G)-nitro-L-arginine methyl ester (L-NAME), an inhibitor of nitric oxide synthase. GSPs suppressed the elevated levels of endogenous NO/NOS in A549 and H1299 cells and blocked the migration promoting capacity of L-arginine. Treatment with guanylate cyclase (GC) inhibitor 1-H-[1,2,4]oxadiaxolo[4,3-a]quinolalin-1-one (ODQ) reduced the migration of A549 cells whereas additional presence of 8-bromoguanosine 3'5'-cyclic monophosphate (8-Br-cGMP, cGMP analogue) restored the migration of these cells, suggesting a role for GC in migration of A549 cells. GSPs reduced the elevated levels of cGMP in cancer cells and also blocked the migration restoring activity of 8-Br-cGMP. The mitogen-activated protein kinase kinase (MAPKK) inhibitor, UO126, inhibited the migration of A549 cells, indicating a role for MAPKK in the migration. Additionally, UO126 and ODQ inhibited the migration restoring effects of L-arginine in L-NAME-treated cells, suggesting the involvement of cGMP and MAPK pathways in NO-mediated migration. GSPs inhibited L-arginine and 8-Br-cGMP-induced activation of ERK1/2 in A549 cells. Together, these results indicate sequential inhibition of NO/NOS, GC, and MAPK pathways by GSPs in mediating the inhibitory signals for cell migration, an essential step in invasion and metastasis.
Collapse
Affiliation(s)
- Thejass Punathil
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Santosh K Katiyar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham VA Medical Center, Birmingham, Alabama
| |
Collapse
|
127
|
Heller A. Apoptosis-inducing high (.)NO concentrations are not sustained either in nascent or in developed cancers. ChemMedChem 2009; 3:1493-9. [PMID: 18759245 DOI: 10.1002/cmdc.200800257] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nitric oxide ((.)NO) induces apoptosis at high concentrations by S-nitrosating proteins such as glyceraldehyde-3-phosphate dehydrogenase. This literature analysis revealed that failure to sustain high (.)NO concentrations is common to all cancers. In cervical, gastric, colorectal, breast, and lung cancer, the cause of this failure is the inadequate expression of inducible nitric oxide synthase (iNOS), resulting from the inhibition of iNOS expression by TGF-beta1 at the mRNA level. In bladder, renal, and prostate cancer, the reason for the insufficient (.)NO levels is the depletion of arginine, resulting from arginase overexpression. Arginase competes with iNOS for arginine, catalyzing its hydrolysis to ornithine and urea. In gliomas and ovarian sarcomas, low (.)NO levels are caused by inhibition of iNOS by N-chlorotaurine, produced by infiltrating neutrophils. Stimulated neutrophils express myeloperoxidase, catalyzing H2O2 oxidation of Cl- to HOCl, which N-chlorinates taurine at its concentration of 19 mM in neutrophils. In squamous cell carcinomas of the skin, ovarian cancers, lymphomas, Hodgkin's disease, and breast cancers, low (.)NO concentrations arise from the inhibition of iNOS by N-bromotaurine, produced by eosinophil-peroxidase-expressing infiltrating eosinophils. Eosinophil peroxidase catalyzes the H2O2 oxidation of Br- to HOBr, which N-brominates taurine to N-bromotaurine at its concentration of 15 mM in eosinophils. In microvascularized tumors, the (.)NO concentration is further depleted; (.)NO is rapidly consumed by red blood cells (RBCs) through S-nitrosation of RBC glutathione and hemoglobin, and by oxidation to nitrate by RBC oxyhemoglobin. Angiogenesis-inhibiting antibodies are currently used to treat cancers; their mode of action is not, as previously thought, reduction of the tumor O2 or nutrient supply. They actually decrease the loss of (.)NO to RBCs.
Collapse
Affiliation(s)
- Adam Heller
- Department of Chemical Engineering, University of Texas, Austin, TX 78712, USA.
| |
Collapse
|
128
|
Abstract
Continuous monitoring of neovascular growth in vivo is required for the development and evaluation of drugs acting as suppressors or stimulators of angiogenesis. The cornea assay consists of the placement of an angiogenesis stimulus (tumor tissue, cell suspension, growth factor) into a micropocket produced in the cornea thickness to evoke vascular outgrowth from the peripherally located limbal vasculature. Neovascular development and progression can be modified by the presence of locally released or applied inhibitory factors or by systemically given antiangiogenic drugs. This assay has the advantage over other in vivo assays of measuring new blood vessels only since the cornea is initially avascular. The experimental details of the avascular cornea assay and its advantages and disadvantages in different species are discussed.
Collapse
Affiliation(s)
- Marina Ziche
- Department of Molecular Biology, University of Siena, Siena, Italy
| | | |
Collapse
|
129
|
Abstract
Cutaneous melanoma preferentially metastasizes via the lymphatic route. However, the mechanisms of lymphatic invasion and metastasis to regional lymph nodes are poorly understood. Nitric oxide is a free radical molecule synthesized from L-arginine by nitric oxide synthases that plays a critical role in various physiological and pathological processes, including tumor growth and angiogenesis. We have tested whether inducible nitric oxide synthase expression correlates with lymphatic vessel density identified with D2-40 antibody and/or blood microvessel density identified with CD105/endoglin in a series of melanocytic nevi (n=28) and cutaneous melanomas (n=38), representative of various pT. Inducible nitric oxide synthase expression was significantly lower in melanocytic nevi in comparison with primary and metastatic melanomas (P<0.001). Mean microvessel density was significantly higher in primary and metastatic melanomas in comparison with melanocytic nevi (P<0.001 for intratumoral and P=0.001 for peritumoral vessels). Vertical growth phase melanomas showed a higher intratumoral microvessel density in comparison with radial growth phase melanomas (P=0.02). The number of peritumoral lymphatics was significantly lower in nevi as compared with primary and metastatic melanomas (P=0.01). No correlation between microvessel or lymphatic vessel and clinical outcome was found in melanomas. A significant direct correlation was observed between inducible nitric oxide synthase immunostaining in melanocytic tumor cells and the density of lymphatic vessels (peritumoral: P=0.001; intratumoral: P=0.08), and the density of peritumoral blood microvessel (P=0.02). Our findings support the hypothesis that inducible nitric oxide synthase is implicated not only in blood, but also in lymphatic vascular neoformation in melanoma. Mechanistic studies are needed to address the possibility that inducible nitric oxide synthase controls lymphangiogenesis, dissemination and lymphatic borne metastases.
Collapse
|
130
|
Yang GY, Taboada S, Liao J. Induced nitric oxide synthase as a major player in the oncogenic transformation of inflamed tissue. Methods Mol Biol 2009; 512:119-156. [PMID: 19347276 DOI: 10.1007/978-1-60327-530-9_8] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nitric oxide (NO) is a free radical that is involved in the inflammatory process and carcinogenesis. There are four nitric oxide synthase enzymes involved in NO production: induced nitric oxide synthase (iNOS), endothelial NO synthase (eNOS), neural NO synthase (nNOS), and mitochondrial NOS. iNOS is an inducible and key enzyme in the inflamed tissue. Recent literatures indicate that NO as well as iNOS and eNOS can modulate cancer-related events including nitro-oxidative stress, apoptosis, cell cycle, angio-genesis, invasion, and metastasis. This chapter focuses on linking NO/iNOS/eNOS to inflammation and carcinogenesis from experimental evidence to potential targets on cancer prevention and treatment.
Collapse
Affiliation(s)
- Guang-Yu Yang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | | |
Collapse
|
131
|
Marangoni K, Araújo TG, Neves AF, Goulart LR. The -786T>C promoter polymorphism of the NOS3 gene is associated with prostate cancer progression. BMC Cancer 2008; 8:273. [PMID: 18823560 PMCID: PMC2571109 DOI: 10.1186/1471-2407-8-273] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 09/29/2008] [Indexed: 11/30/2022] Open
Abstract
Background There is no biological or epidemiological data on the association between NOS3 promoter polymorphisms and prostate cancer. The polymorphisms in the promoter region of NOS3 gene may be responsible for variations in the plasma NO, which may promote cancer progression by providing a selective growth advantage to tumor cells by angiogenic stimulus and by direct DNA damage. Methods This study aimed evaluating the NOS3 promoter polymorphisms by PCR-SSCP and sequencing, associating genotypes and haplotypes with NOS3 expression levels through semi-quantitative RT-PCR, and with PCA3 mRNA detection, a specific tumor biomarker, in the peripheral blood of pre-surgical samples from 177 patients; 83 PCa and 94 BPH. Results Three novel SNPs were identified -764A>G, -714G>T and -649G>A in the NOS3 gene promoter region, which together with the -786T>C generated four haplotypes (N, T, C, A). NOS3 gene expression levels were affected by the -786T>C polymorphism, and there was a 2-fold increase in NOS3 levels favored by the incorporation of each C allele. NOS3 levels higher than 80% of the constitutive gene expression level (B2M) presented a 4-fold increase in PCa occurrence. Conclusion The -786T>C polymorphism was the most important promoter alteration of the NOS3 gene that may affect the PCa progression, but not its occurrence, and the incorporation of the C allele is associated with increased levels of NOS3 transcripts. The NOS3 transcript levels presented a bimodal behavior in tumor development and may be used as a biomarker together with the PCA3 marker for molecular staging of the prostate cancer.
Collapse
Affiliation(s)
- Karina Marangoni
- Federal University of Uberlândia, Institute of Genetics and Biochemistry, Molecular Genetics Laboratory, Campus Umuarama, Block 2E, Room 24, 38400-902, Uberlândia, MG, Brazil.
| | | | | | | |
Collapse
|
132
|
Chen JH, Lin HH, Chiang TA, Hsu JD, Ho HH, Lee YC, Wang CJ. Gaseous nitrogen oxide promotes human lung cancer cell line A549 migration, invasion, and metastasis via iNOS-mediated MMP-2 production. Toxicol Sci 2008; 106:364-75. [PMID: 18796497 DOI: 10.1093/toxsci/kfn195] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Gaseous nitrogen oxide (gNO) is an important indoor and outdoor air pollutant. Many studies have indicated gNO causes lung tissue damage by its oxidation properties and free radicals. However, there are considerably few data on the association between lung cancer and gNO exposure. The purpose of this study was to examine whether gNO could contribute to the process of malignant progression of lung cancer. The results of wound-healing assay and in vitro transwell assay revealed that gNO-induced dose and time dependently the migration and invasion of A549 cells, a human lung cancer cell line, under noncytotoxic concentrations. gNO was able to induce release of NO from A549 cells, an effect that was mediated via the activation of inducible nitric oxide synthases (iNOS), but not constitutive isoforms, during the same treatment period. An increased expression of matrix metalloproteinase (MMP) and a coincided reduction in repress tissue inhibitors of metalloprotease-2 were observed upon the treatment of gNO. The gNO-mediated MMP-2 induction appeared to be a consequence of nuclear factor kappa B and activation protein-1 activation, because that their DNA binding activity was enhanced by gNO. All these influences of gNO were efficiently repressed by the pretreatment of a NOS inhibitor (N(G)-nitro-L-arginine methyl ester). Using a mouse model, we showed that gNO promoted A549 metastasis to the lung through a mechanism involving the iNOS-dependent MMP-2 activity. Our data imply that gNO exposure, which in turn led to iNOS activation and the enhancement of MMP-mediated cellular events, was related to lung cancer development.
Collapse
Affiliation(s)
- Jing-Hsien Chen
- Graduate Institute of Biological Science and Technology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
133
|
Mikirova NA, Ichim TE, Riordan NH. Anti-angiogenic effect of high doses of ascorbic acid. J Transl Med 2008; 6:50. [PMID: 18789157 PMCID: PMC2562367 DOI: 10.1186/1479-5876-6-50] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 09/12/2008] [Indexed: 12/14/2022] Open
Abstract
Pharmaceutical doses of ascorbic acid (AA, vitamin C, or its salts) have been reported to exert anticancer activity in vitro and in vivo. One proposed mechanism involves direct cytotoxicity mediated by accumulation of ascorbic acid radicals and hydrogen peroxide in the extracellular environment of tumor cells. However, therapeutic effects have been reported at concentrations insufficient to induce direct tumor cell death. We hypothesized that AA may exert anti-angiogenic effects. To test this, we expanded endothelial progenitor cells (EPCs) from peripheral blood and assessed, whether or not high dose AA would inhibit EPC ability to migrate, change energy metabolism, and tube formation ability. We also evaluated the effects of high dose AA on angiogenic activities of HUVECs (human umbilical vein endothelial cells) and HUAECs (human umbilical arterial endothelial cells). According to our data, concentrations of AA higher than 100 mg/dl suppressed capillary-like tube formation on Matrigel for all cells tested and the effect was more pronounced for progenitor cells in comparison with mature cells. Co-culture of differentiated endothelial cells with progenitor cells showed that there was incorporation of EPCs in vessels formed by HUVECs and HUAECs. Cell migration was assessed using an in vitro wound healing model. The results of these experiments showed an inverse correlation between AA concentrations relative to both cell migration and gap filling capacity. Suppression of NO (nitric oxide) generation appeared to be one of the mechanisms by which AA mediated angiostatic effects. This study supports further investigation into non-cytotoxic antitumor activities of AA.
Collapse
Affiliation(s)
- Nina A Mikirova
- Bio-Communications Research Institute, Wichita, Kansas, USA.
| | | | | |
Collapse
|
134
|
Effect of using RNA interference to alter iNOS gene expression on the proliferation of tongue squamous cell carcinoma cell line Tca8113. Br J Oral Maxillofac Surg 2008; 46:435-8. [DOI: 10.1016/j.bjoms.2008.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2008] [Indexed: 11/17/2022]
|
135
|
Jayakumar R, Sasikala K. Evaluation of DNA damage in jewellery workers occupationally exposed to nitric oxide. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2008; 26:259-261. [PMID: 21783919 DOI: 10.1016/j.etap.2008.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 03/26/2008] [Accepted: 03/29/2008] [Indexed: 05/31/2023]
Abstract
Nitric oxide is a metastable radical, reacts with oxygen to produce toxic nitrogen oxides (N(2)O(3), ONOO(-)) which damage DNA. Occupational exposure to nitric oxide leads to increased frequency of chromosomal aberrations in humans. In the present study the DNA damage among the jewellery workers occupationally exposed to nitric oxide was analyzed using buccal cell comet assay. The result of this study shows increased levels of DNA damage among jewellery workers. The habit of cigarette smoking among the jewellery workers has a synergistic effect on inducing DNA damage.
Collapse
Affiliation(s)
- Rajarajeswaran Jayakumar
- Unit of Human Genetics, Department of Zoology, Bharathiar University, Coimbatore 641046, Tamil nadu, India
| | | |
Collapse
|
136
|
Heterogeneous nuclear ribonucleoprotein G, nitric oxide, and oral carcinogenesis. Nitric Oxide 2008; 19:125-32. [DOI: 10.1016/j.niox.2008.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/12/2008] [Accepted: 04/16/2008] [Indexed: 11/23/2022]
|
137
|
Punathil T, Tollefsbol TO, Katiyar SK. EGCG inhibits mammary cancer cell migration through inhibition of nitric oxide synthase and guanylate cyclase. Biochem Biophys Res Commun 2008; 375:162-7. [PMID: 18692479 DOI: 10.1016/j.bbrc.2008.07.157] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Accepted: 07/30/2008] [Indexed: 11/15/2022]
Abstract
Tumor cell migration is considered as a major event in the metastatic cascade. Here we examined the effect of (-)-epigallocatechin-3-gallate (EGCG) on migration capacity and molecular mechanism using 4T1 murine mammary cancer cells as a model. Using an in vitro migration assay, we found that treatment of 4T1 cells with EGCG resulted in concentration-dependent inhibition of migration of these cells. The migration capacity of cells was reduced in presence of N(G)-nitro-l-arginine methyl ester (l-NAME), an inhibitor of nitric oxide synthase. EGCG suppressed the elevated levels of endogenous NO/NOS in 4T1 cells and blocked the migration promoting capacity of l-arginine. Treatment with guanylate cyclase inhibitor 1-H-[1,2,4]oxadiaxolo[4,3-a]quinolalin-1-one (ODQ) reduced the migration of 4T1 cells. EGCG reduced the elevated levels of cGMP in cancer cells and blocked the migration restoring activity of 8-Br cGMP (cGMP analogue). These results indicate for the first time that EGCG inhibits mammary cancer cell migration through the inhibition of NO/NOS and guanylate cyclase.
Collapse
Affiliation(s)
- Thejass Punathil
- Department of Dermatology, University of Alabama at Birmingham, 1670 University Boulevard, Volker Hall 557, Birmingham, AL, USA
| | | | | |
Collapse
|
138
|
|
139
|
Fan CL, Zhou JH, Hu CY. Expression of human papillomavirus in mammary carcinoma and its possible mechanism in carcinogenesis. Virol Sin 2008. [DOI: 10.1007/s12250-008-2914-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
140
|
Guruvayoorappan C, Kuttan G. (+)-Catechin inhibits tumour angiogenesis and regulates the production of nitric oxide and TNF-α in LPS-stimulated macrophages. Innate Immun 2008; 14:160-74. [DOI: 10.1177/1753425908093295] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The anti-angiogenic activity of (+)-catechin as well as its regulatory effect on the production of nitric oxide and TNFα were studied using in vivo and in vitro models. In vivo angiogenic activity was studied using B16F-10 melanoma cell-induced capillary formation in C57BL/6 mice. Administration of (+)-catechin significantly inhibited (36.09%) the number of tumour-directed capillaries induced by injecting B16F-10 melanoma cells on the ventral side of C57BL/6 mice. The cytokine profile in the serum of these animals showed a drastically increased level of proinflammatory cytokines such as IL-1β, IL-6, TNF-α, GM-CSF and the direct endothelial cell proliferating agent, VEGF. Administration of (+)-catechin could differentially regulate elevation of these cytokines. The differential elevation is further evidenced by the increased production of IL-2 and tissue inhibitor of metalloproteinase-1 (TIMP-1) in the B16F-10 injected, (+)-catechin-treated animals. In vitro L929 bioassay revealed the inhibition of TNF-α production by (+)-catechin treatment. In the rat aortic ring assay, (+)-catechin inhibited the microvessel outgrowth at non-toxic concentrations. (+)-Catechin at non-toxic concentrations (5—25 µg/ml) showed significant inhibition in the proliferation, migration and tube formation of endothelial cells, which are the key events in the process of angiogenesis. (+)-Catechin also showed inhibitory effect on VEGF mRNA levels in B16F-10 melanoma cells. (+)-Catechin inhibited the production of NO and TNF-α in LPS-stimulated primary macrophages. Taken together, these results demonstrate that (+)-catechin inhibits tumour-specific angiogenesis by regulating the production of pro- and anti-angiogenic factors such as pro-inflammatory cytokines, nitric oxide, VEGF, IL-2 and TIMP-1. These results also suggest that (+)-catechin could significantly inhibit nitrite and TNF-α production in LPS-stimulated macrophages.
Collapse
Affiliation(s)
| | - Girija Kuttan
- Department of Immunology, Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India,
| |
Collapse
|
141
|
The low molecular weight S-nitrosothiol, S-nitroso-N-acetylpenicillamine, promotes cell cycle progression in rabbit aortic endothelial cells. Nitric Oxide 2008; 18:241-55. [DOI: 10.1016/j.niox.2008.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Revised: 01/31/2008] [Accepted: 02/03/2008] [Indexed: 11/23/2022]
|
142
|
iNOS as a therapeutic target for treatment of human tumors. Nitric Oxide 2008; 19:217-24. [PMID: 18515106 DOI: 10.1016/j.niox.2008.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 05/02/2008] [Accepted: 05/05/2008] [Indexed: 01/25/2023]
Abstract
Nitric oxide synthase (NOS) has been shown to be overexpressed in a number of human tumors compared to normal tissues and therefore potentially represents an exploitable target in future anticancer therapies. To achieve this, there will be a need to profile tumors to identify those expressing high levels of NOS; alternatively, endogenous (low) levels of NOS could be modulated by induction or through gene therapy approaches. NOS consists of a reductase domain which shares a high degree of sequence homology with P450 reductase and this domain supplies reducing equivalents to a haem containing oxygenase domain that is responsible for the production of nitric oxide. Thus, there are a number of routes of exploitation. Firstly, to take advantage of the reductase domain to activate bioreductive drugs as has been exemplified with tirapazamine and now extended to AQ4N (1,4-bis{2-(dimethylamino-N-oxide)ethylamino}5,8-dihydroxy-anthracene-9,10-dione). Secondly, to take advantage of nitric oxide production for its ability to increase the sensitivity of resistant hypoxic cells to radiation. Lastly, to utilize inhibition of HIF-1 to amplify NO based therapies. In this review we provide examples/evidence of how these objectives can be achieved.
Collapse
|
143
|
Yasuda H. Solid tumor physiology and hypoxia-induced chemo/radio-resistance: novel strategy for cancer therapy: nitric oxide donor as a therapeutic enhancer. Nitric Oxide 2008; 19:205-16. [PMID: 18503779 DOI: 10.1016/j.niox.2008.04.026] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Revised: 04/27/2008] [Accepted: 04/28/2008] [Indexed: 11/30/2022]
Abstract
Hypoxia exists in solid tumor tissues due to abnormal vasculature, vascular insufficiency, treatment or malignancy related anemia, and low intratumor blood flow. Hypoxic status in solid tumor promotes accumulation of hypoxia-inducible factor-1 alpha which is promptly degraded by proteasomal ubiquitination under normoxic conditions. However, under hypoxic conditions, the ubiquitination system for HIF-1 alpha is inhibited by inactivation of prolyl hydroxylase which is responsible for hydroxylation of proline in the oxygen-dependent degradation domain of HIF-1 alpha. HIF-1 alpha is an important transcriptional factor that codes for hundreds of genes involved in erythropoiesis, angiogenesis, induction of glycolytic enzymes in tumor tissues, modulation of cancer cell cycle, cancer proliferation, and cancer metastasis. Hypoxia and accumulation of HIF-1 alpha in solid tumor tissues have been reported to associate with resistance to chemotherapy, radiotherapy, and immunotherapy and poor prognosis. Production of vascular endothelial growth factor (VEGF) in cancer cells is regulated by the activated HIF-1 mediated system. An increase in VEGF levels subsequently induces HIF-1 alpha accumulation and promotes tumor metastasis by angiogenesis. Recently, angiogenesis targeting therapy using humanized VEGF antibody and VEGF receptor tyrosine kinase inhibitors have been used in solid cancer therapy. Nitric oxide (NO) is a unique chemical gaseous molecule that plays a role as a chemical messenger involved in vasodilator, neurotransmitter, and anti-platelet aggregation. In vivo, NO is produced and released from three different isoforms of NO synthase (NOS) and from exogenously administered NO donors. In cancer science, NO has been mainly discussed as an oncogenic molecule over the past decades. However, NO has recently been noted in cancer biology associated with cancer cell apoptosis, cancer cell cycle, cancer progression and metastasis, cancer angiogenesis, cancer chemoprevention, and modulator for chemo/radio/immuno-therapy. The presence and activities of all the three isoforms of NOS and were detected in cancer tissue components such as cancer cells, tumor-associated macrophages, and vascular endothelium. Overexpression of iNOS in cancer tissues has been reported to associate with poor prognosis in patients with cancers. On the other hand, NO donors such as nitroglycerin have been demonstrated to improve the effects of cancer therapy in solid cancers. Nitroglycerin has been used safely for a long time as a potent vasodilator for the treatment of ischemic heart diseases or heart failure. Therefore, we think highly of clinical use of nitroglycerin as a novel cancer therapy in combination with anticancer drugs for improvement of cancer therapeutic levels. In this review article, we demonstrate the unique physiological characteristics of malignant solid tumors, several factors in solid tumors resulting in resistance for cancer therapies, and the effects of NO from NOS or exogenous NO-donating drugs on malignant cells. Furthermore, we refer to promising therapeutic roles of NO and NO-donating drugs for novel treatments in solid tumors.
Collapse
Affiliation(s)
- Hiroyasu Yasuda
- Department of Translational Clinical Oncology, Kyoto University Graduate School of Medicine, Kyoto University Hospital, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
144
|
Brennan PA, Dennis S, Poller D, Quintero M, Puxeddu R, Thomas GJ. Inducible nitric oxide synthase: correlation with extracapsular spread and enhancement of tumor cell invasion in head and neck squamous cell carcinoma. Head Neck 2008; 30:208-14. [PMID: 17657783 DOI: 10.1002/hed.20675] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Extracapsular nodal spread is a major prognostic indicator in head and neck cancer. Nitric oxide (NO), primarily produced by the enzyme inducible NO synthase (iNOS), has a large number of actions in cancer biology, but no studies have investigated its possible role in extracapsular spread or tumor invasion. METHODS Immunochemistry was used to study iNOS expression in 48 patients with either extracapsular or encapsulated metastasis. In vitro invasion assays were performed using H357 (an oral squamous cell carcinoma cell line) using the iNOS inhibitor drug, 1400 W. RESULTS iNOS expression was significantly associated with extracapsular spread, with 22/27 cases showing positive iNOS expression compared with 8/21 cases in the encapsulated group (p = .01). Invasion of H357 cells was inhibited by 1400 W at concentrations of 100 microM and 1 mM (p = .002, p = .003). CONCLUSION iNOS protein seems to be associated with extracapsular spread and invasion in head and neck cancer. Further studies are required to understand this role more fully.
Collapse
Affiliation(s)
- Peter A Brennan
- Department of Maxillofacial Head and Neck Surgery, Queen Alexandra Hospital, Portsmouth PO6 3LY, United Kingdom.
| | | | | | | | | | | |
Collapse
|
145
|
Han EH, Hwang YP, Lee KJ, Jeong TC, Jeong HG. 1-Bromopropane induces macrophage activation via extracellular signal-regulated kinase 1/2 MAPK and NF-κB pathways. Cancer Lett 2008; 262:28-36. [DOI: 10.1016/j.canlet.2007.11.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Revised: 11/20/2007] [Accepted: 11/20/2007] [Indexed: 11/28/2022]
|
146
|
Pluta RM. Dysfunction of nitric oxide synthases as a cause and therapeutic target in delayed cerebral vasospasm after SAH. ACTA NEUROCHIRURGICA. SUPPLEMENT 2008; 104:139-47. [PMID: 18456999 PMCID: PMC4762030 DOI: 10.1007/978-3-211-75718-5_28] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nitric oxide (NO), also known as endothelium-derived relaxing factor, is produced by endothelial nitric oxide synthase (eNOS) in the intima and by neuronal nitric oxide synthase (nNOS) in the adventitia of cerebral vessels. It dilates the arteries in response to shear stress, metabolic demands, pterygopalatine ganglion stimulation, and chemoregulation. Subarachnoid haemorrhage (SAH) interrupts this regulation of cerebral blood flow. Hemoglobin, gradually released from erythrocytes in the subarachnoid space destroys nNOS-containing neurons in the conductive arteries. This deprives the arteries of NO, leading to the initiation of delayed vasospasm. But such vessel narrowing increases shear stress, which stimulates eNOS. This mechanism normally would lead to increased production of NO and dilation of arteries. However, a transient eNOS dysfunction evoked by an increase of the endogenous competitive nitric oxide synthase (NOS) inhibitor, asymmetric dimethyl-arginine (ADMA), prevents this vasodilation. eNOS dysfunction has been recently shown to be evoked by increased levels of ADMA in CSF in response to the presence of bilirubin-oxidized fragments (BOXes). A direct cause of the increased ADMA CSF level is most likely decreased ADMA elimination due to the disappearance of ADMA-hydrolyzing enzyme (DDAH II) immunoreactivity in the arteries in spasm. This eNOS dysfunction sustains vasospasm. CSF ADMA levels are closely associated with the degree and time-course of vasospasm; when CSF ADMA levels decrease, vasospasm resolves. Thus, the exogenous delivery of NO, inhibiting the L-arginine-methylating enzyme (IPRMT3) or stimulating DDAH II, may provide new therapeutic modalities to prevent and treat vasospasm. This paper will present results of preclinical studies supporting the NO-based hypothesis of delayed cerebral vasospasm development and its prevention by increased NO availability.
Collapse
Affiliation(s)
- R M Pluta
- Surgical Neurology Branch, National Institutes of Health, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA.
| |
Collapse
|
147
|
Morbidelli L, Donnini S, Ziche M. Nitric Oxide in Tumor Angiogenesis. Angiogenesis 2008. [DOI: 10.1007/978-0-387-71518-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
148
|
Yamaguchi H, Noshita T, Kidachi Y, Umetsu H, Hayashi M, Komiyama K, Funayama S, Ryoyama K. Isolation of Ursolic Acid from Apple Peels and Its Specific Efficacy as a Potent Antitumor Agent. ACTA ACUST UNITED AC 2008. [DOI: 10.1248/jhs.54.654] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | - Toshiro Noshita
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| | - Yumi Kidachi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| | - Hironori Umetsu
- Laboratory of Food Chemistry, Department of Life Sciences, Junior College, Gifu Shotoku Gakuen University
| | | | | | - Shinji Funayama
- Department of Kampo Pharmaceutical Sciences, Nihon Pharmaceutical University
| | - Kazuo Ryoyama
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Aomori University
| |
Collapse
|
149
|
Segawa Y, Oda Y, Yamamoto H, Uryu H, Shiratsuchi H, Hirakawa N, Tomita K, Yamamoto T, Oda S, Yamada T, Komune S, Tsuneyoshi M. Overexpression of inducible nitric oxide synthase and accumulation of 8-OHdG in nasopharyngeal carcinoma. Histopathology 2007; 52:213-23. [DOI: 10.1111/j.1365-2559.2007.02920.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
150
|
Shigyo H, Nonaka S, Katada A, Bandoh N, Ogino T, Katayama A, Takahara M, Hayashi T, Harabuchi Y. Inducible nitric oxide synthase expression in various laryngeal lesions in relation to carcinogenesis, angiogenesis, and patients' prognosis. Acta Otolaryngol 2007; 127:970-9. [PMID: 17712678 DOI: 10.1080/00016480601089382] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
CONCLUSIONS The inducible nitric oxide synthase (iNOS) expression leading to vascular endothelial growth factor (VEGF) overexpression may be useful as a factor for predicting recurrence after initial treatment and prognosis in laryngeal squamous cell carcinoma (SCC). OBJECTIVE We analyzed expression of iNOS, p53, and VEGF in various laryngeal lesions. MATERIALS AND METHODS The study samples consisted of 63 SCC, 20 dysplasia, 7 polyp, and 5 normal epithelium of the larynx. The expression of iNOS, p53, and VEGF was identified by immunohistological methods. RESULTS No positive immunostaining for iNOS, p53, and VEGF was observed in normal epithelium and polyps. In contrast, with the progression from mild/moderate dysplasia to severe dysplasia to carcinoma, their expression levels increased. In dysplasia, there was a significant positive correlation among expression of iNOS, p53, and VEGF. In SCC, iNOS expression correlated with VEGF overexpression and microvessel density, but not with p53 overexpression. In SCC, the expression of iNOS and VEGF significantly increased in patients who developed local recurrence and/or metastases after initial treatments. Kaplan-Meier analysis showed that disease-free survival was significantly shorter in patients with iNOS or VEGF expression. Multivariate analysis showed expression of iNOS and VEGF as independent indicators for poor disease-free survival.
Collapse
Affiliation(s)
- Hiroshi Shigyo
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical College, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|