High concentrations of nitric oxide (NO) regulation of NO synthase (NOS) activity is essential for minimizing effects of cytotoxic and genotoxic nitrogen oxide species. We have shown previously, that NO-induced p53 protein accumulation, down-regulates basal and cytokine-modulated inducible NOS (NOS2) expression in human cells in vitro, and that p53-null mice have elevated NOS2 enzymatic activity. Our investigation of primary colon tumors establishes a strong positive relationship between the presence of NOS2 in tumors and the frequency of G:C to A:T transitions at CpG dinucleotides. These mutations also are common in lymphoid, esophageal, head and neck, stomach, brain and breast cancers. Increased NOS2 expression has been demonstrated in four of these cancers. Tumor-associated NO production may modify DNA directly, or may inhibit DNA repair activities, such as the recently described human thymine-DNA glycosylase, which has been shown to repair G:T mismatches at CpG dinucleotides. Because NO production also induces the accumulation of wild-type p53, the resulting growth inhibition can provide an additional strong selection pressure for nonfunctional, mutant p53. NO may, therefore, act as both an endogenous initiator and promoter in human colon carcinogenesis, and specific inhibitors of NOS2, as demonstrated recently in an animal tumor model, may have important chemopreventive potential in human colorectal cancer. These and other findings indicate that NO has a pathophysiological role in carcinogenesis. To determine the role of NO in tumor progression, we generated human carcinoma cell lines that produced NO constitutively. Cancer cells expressing NOS2 that had wild-type p53, had reduced tumor growth in athymic nude mice, whereas those with mutated p53 had accelerated tumor growth associated with increased vascular endothelial growth factor expression and neovascularization. Our data indicate that tumor-associated NO production may promote cancer progression by providing a selective growth advantage to tumor cells with mutant p53, and that inhibitors of NOS2 may have therapeutic activity in these tumors. We are also investigating chronic inflammatory diseases, e.g., ulcerative colitis, and genetic oxyradical overload diseases, e.g., hemochromatosis and Wilson Disease, that are cancer prone. p53 mutation load is increased in these cancer-prone conditions. NOS2 and COX2 are increased in a portion of the cases. NOS2 expression can be regulated by the WNT-APC-b-catenin pathway. Nitric oxide activates p53 by its post-translation by serine kinases. Currently, we are investigating the interaction of the NOS2 and COX2 pathways.

Agency
National Institute of Health (NIH)
Institute
Division of Basic Sciences - NCI (NCI)
Type
Intramural Research (Z01)
Project #
1Z01BC010033-05
Application #
6433193
Study Section
(LHC)
Project Start
Project End
Budget Start
Budget End
Support Year
5
Fiscal Year
2000
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Stauffer, Jimmy K; Scarzello, Anthony J; Jiang, Qun et al. (2012) Chronic inflammation, immune escape, and oncogenesis in the liver: a unique neighborhood for novel intersections. Hepatology 56:1567-74
Simone, Nicole L; Soule, Benjamin P; Ly, David et al. (2009) Ionizing radiation-induced oxidative stress alters miRNA expression. PLoS One 4:e6377
Wink, David A; Ridnour, Lisa A; Hussain, S Perwez et al. (2008) The reemergence of nitric oxide and cancer. Nitric Oxide 19:65-7
Hussain, S Perwez; He, Peijun; Subleski, Jeffery et al. (2008) Nitric oxide is a key component in inflammation-accelerated tumorigenesis. Cancer Res 68:7130-6
Thomas, Douglas D; Ridnour, Lisa A; Isenberg, Jeffrey S et al. (2008) The chemical biology of nitric oxide: implications in cellular signaling. Free Radic Biol Med 45:18-31
Thomas, Douglas D; Ridnour, Lisa A; Espey, Michael Graham et al. (2006) Superoxide fluxes limit nitric oxide-induced signaling. J Biol Chem 281:25984-93
Goodman, Julie E; Mechanic, Leah E; Luke, Brian T et al. (2006) Exploring SNP-SNP interactions and colon cancer risk using polymorphism interaction analysis. Int J Cancer 118:1790-7
Ying, Lei; Marino, Jillian; Hussain, S Perwez et al. (2005) Chronic inflammation promotes retinoblastoma protein hyperphosphorylation and E2F1 activation. Cancer Res 65:9132-6
Hofseth, Lorne J; Robles, Ana I; Espey, Michael G et al. (2005) Nitric oxide is a signaling molecule that regulates gene expression. Methods Enzymol 396:326-40
Hussain, S Perwez; Trivers, Glennwood E; Hofseth, Lorne J et al. (2004) Nitric oxide, a mediator of inflammation, suppresses tumorigenesis. Cancer Res 64:6849-53

Showing the most recent 10 out of 29 publications