The adaptive imbalance to genotoxic stress: genome guardians rear their ugly heads

Lorne J. Hofseth

Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA

Email: hofseth{at}cop.sc.edu


    Abstract
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
An adaptive response of the genome-protection machinery occurs in cells exposed to genotoxic stress. This machinery includes the p53 and retinoblastoma protein pathways, which are not mutually exclusive from other adapting machinery including DNA repair, cell cycle checkpoints, apoptosis and endogenous metabolizing and antioxidant enzymes. The adaptive changes occur in chronic inflammation and in cigarette smokers associated with a high cancer risk, and are an attempt to keep cells healthy. However, there is increasing evidence that this response may have deleterious effects. Here, key pathways that adaptively respond to genotoxic stress are reviewed and mechanisms by which this response may have pro-carcinogenic implications are discussed.

Abbreviations: BER, base excision repair; Gadd, growth arrest and DNA damage-inducible; GPx, glutathione peroxidase; NO, nitric oxide; pRb, retinoblastoma protein; SOD, superoxide dismutase


    Introduction
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
When a cell is exposed to genotoxic stress through viral, chemical or physical means, an adaptive response occurs involving an adjustment in the levels and/or activity of its genome-protecting protein machinery. These proteins are involved in DNA repair, cell cycle checkpoints and apoptosis. Some have free radical scavenging properties (Table I). This is a survival mechanism aimed at maintaining good cellular health. The following is a discussion of the current knowledge of proteins and pathways that adaptively respond to genotoxic stress. Highlighted at the end of each section is the emerging evidence that these adaptive responses can paradoxically generate genotoxic stress and exacerbate carcinogenesis. A summary of these pro-carcinogenic mechanisms of genome guardian adaptive responses is provided in Table II.


View this table:
[in this window]
[in a new window]
 
Table I. Examples of mammalian genome protecting proteins that are induced in vitro and in vivo by genotoxic stress

 

View this table:
[in this window]
[in a new window]
 
Table II. Examples of pro-carcinogenic characteristics of genome guardians

 

    p53 adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
The p53 protein is a key tumor suppressor protein that lies at the crossroads of cellular stress response pathways (1,2). Through these pathways resulting in cell cycle checkpoint arrest, DNA repair, cellular senescence, differentiation, apoptosis and free radical generation, p53 facilitates the repair and survival of damaged cells or eliminates severely damaged cells from the replicative pool to protect the organism. This p53 adaptive response was first observed with UV (3) and {lambda}-IR (4,5), and has since been shown to occur with many other genotoxic stresses, including chronic inflammation (6).

For the most part, induction of p53 has a favorable outcome to the organism, and thus a primary focus for cancer treatment is the induction and stabilization of wild-type p53. However, this induction may not have favorable effects if there is an induction of a mutated form of p53. Proof-of-principle comes from mouse experiments where transgenic mice carrying mutant p53 have accelerated tumorigenesis (711). Interestingly, p53 mutant mice have an augmented p53 response to genotoxic stress (12). This leads to speculation that this may occur in patients with high cancer risk, chronic inflammatory (‘oxyradical overload’) diseases, because these patients have a high p53 mutation load (13,14) with inducible p53 (6). Interestingly, p53 has also been shown to transactivate genes involved in free radical formation (15,16). Although pro-oxidant states (17) are pro-apoptotic, they may have deleterious effects on the cell by generating damage in key cancer genes and proteins.

Another key tumor suppressor protein deserving attention here is the retinoblastoma protein (pRb). pRb levels have been shown to be elevated in colorectal carcinomas. This apparent paradox has been discussed previously (18). Also worth mentioning is the issue of pRb and its functional dependence on phosphorylation status (19). In a pRb hyperphosphorylated state, E2F transcription factors are released, which activates the DNA replication machinery and allows cell cycle progression. Therefore, a careful assessment of pRb phosphorylation status is necessary to rule out that the increase in pRb protein levels is not associated with a concomitant increase in pRb phosphorylation. If so, then this could explain the apparent transformation of this genome guardian to that of a pro-carcinogenic molecule.


    DNA repair adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
It has long been known that bacteria have an SOS response, in which DNA repair is induced following DNA damage (20,21). Although less robust, the repair machinery of mammalian cells can also adapt to genotoxic stress. For example, ß-polymerase and 3-methyl-adenine DNA glycosylase (AAG), two enzymes involved in base excision repair (BER), and O6-methylguanine-DNA methyltransferase, are inducible by genotoxic stress (2224). More recently, studies have found the DNA repair machinery adaptively responds to oxidative and nitrosative stress, both in vitro and in vivo (2529). This response is an attempt to repair DNA damage created by the genotoxic stress and maintain good cellular health. Evidence is now emerging that this response could have deleterious effects.

The pro-mutagenic consequences of DNA repair was first identified in the Escherichia coli SOS repair mechanism. In the early 1990 s, Xiao and Samson demonstrated that imbalanced BER could affect spontaneous mutation rates (30). Recent studies have shown that over-expression and an imbalance in mismatch repair proteins can lead to increased mutation rates (31,32). In mammalian cells, over-expression of specific BER enzymes generates chromosomal instability and may contribute to tumorigenesis (33,34). We have shown that there is an imbalance in BER enzymes in chronic inflammation and that this could generate microsatellite instability in patients with the high colon cancer risk, oxyradical overload disease ulcerative colitis (29).


    Cell cycle checkpoint adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
Genotoxic stress in cells leads to a temporary growth arrest. This pause, or cell cycle arrest, bides time for repair of damaged cellular DNA before DNA replication occurs. Critical molecules involved in cell cycle checkpoints are cyclins and their associated kinases (cyclin-dependent kinases). Negative regulators of cyclins and cyclin-dependent kinases can act as a braking mechanism and pause the cell in the G1, S or G2 phase of the cell cycle. The G1 delay allows for repair of damage before it is replicated. The S phase completion checkpoint, activated by stalled replication machinery and DNA damage, and the G2 checkpoint have a protective effect by allowing additional time for repair of DNA damage prior to entry into mitosis (35). p21Waf1/Cip1/Sdi1 is a key protein involved in both G1 (36) and G2 (37) cell cycle arrest. Gadd45 (growth arrest and DNA damage-inducible) is a key protein involved in a G2 cell cycle arrest (38). Both these proteins are inducible and have been shown to be elevated in chronic inflammation (6,39,40).

There is some evidence that expression of p21Waf1/Cip1/Sdi1 or Gadd45 has negative effects on genomic stability. Smith et al. (41,42) found a lack of Gadd45 resulted in an increased sensitivity to cell killing by UV radiation or cisplatin, indicating a possibility that over-expression of Gadd45 leads to reduced cell mortality with carcinogenic consequences. As a cell cycle checkpoint protein, p21Waf1/Cip1/Sdi1 plays a key role in tumor suppression. However, there is also evidence that p21Waf1/Cip1/Sdi1 has genome destabilizing and oncogenic functions (43). Similar to Gadd45, loss of p21Waf1/Cip1/Sdi1 renders cells sensitive to ionizing radiation (44). This loss also delays the onset of lymphoma in ATM-deficient mice (44). Other studies have reported that anti-sense knockdown of p21Waf1/Cip1/Sdi1 inhibits cyclin D complex assembly and growth factor-mediated cellular proliferation (45). Recently, Dong et al. (46) showed stimulation of proliferation with p21Waf1/Cip1/Sdi1 over-expression. Although some studies indicate a positive correlation of p21Waf1/Cip1/Sdi1 expression with a positive prognosis (4752), other studies find a negative association between p21Waf1/Cip1/Sdi1 expression and prognosis (5358). Several mechanistic studies could explain these observations of a pro-carcinogenic role for p21Waf1/Cip1/Sdi1. p21Waf1/Cip1/Sdi1 can generate endoreduplication, which is a doubling of DNA content due to an unscheduled round of DNA replication (59,60). Studies have also shown that following p21Waf1/Cip1/Sdi1-induced growth arrest, abnormal mitosis develops in tumor cells (61,62). Wild-type p21Waf1/Cip1/Sdi1 can also inhibit apoptosis and stimulate transcription of secreted factors with mitogenic and anti-apoptotic acitivites (43). These studies then highlight a pro-carcinogenic role for p21Waf1/Cip1/Sdi1 and a need to reassess the role of this protein as a genome protector.


    Apoptosis adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
Apoptosis can occur if a stress is so genotoxic that repair is not an option. Insufficient apoptosis can manifest as cancer or autoimmunity, while accelerated cell death is evident in acute and chronic degenerative diseases, immunodeficiency and infertility (63). Two main apoptotic signaling pathways have been identified: a death receptor-dependent (extrinsic) pathway and a mitochondrion-dependent (intrinsic) pathway. In the intrinsic apoptotic pathway, the mitochondrion harbors both anti-apoptotic (Bcl-2, Bcl-XL) and pro-apoptotic factors (Smac/Diablo, Apaf-1, cytochrome c, bax, AIF). Cytochrome c, one of these apoptotic factors, is a respiratory chain component that, when expelled to the cytoplasm during the apoptotic chain of events, assembles with two other molecules (Apaf-1 and procaspase 9) to form the apoptosome. This, then, starts apoptosis execution. Some of the stress-inducible players in both pathways include BH3-only molecules (64,65), caspases (66,67) and death receptors (68,69). Because anti-apoptosis molecules (e.g. bcl-2, inhibitors of apoptosis) can be simultaneously down-regulated by genotoxic stress, the ratio of anti- to pro-apoptotic molecules (e.g. bcl-2/bax) can drive a cell into apoptosis. Therefore, similar to the BER pathway, a balance between enzymes within a pathway can determine whether a cell lives or dies. This adaptive imbalance resulting in cell survival might have deleterious effects because this allows for a cell to accumulate genetic mutations and protein damage.


    Antioxidant adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
An adaptive response of antioxidant defences often occurs in cells exposed to oxidative and nitrosative stress. Enzymes critical to this process include catalase, superoxide dismutase (SOD) and glutathione peroxidase (GPx). SOD catalyzes the dismutation of superoxide radicals to hydrogen peroxide, which is further metabolized to water and oxygen by catalase and GPx (70) (Figure 1). Another enzyme with antioxidant properties, the mechanisms of which are not entirely understood, is hemeoxygenase-1 (HO-1). Although many studies using animal models of inflammation or examining oxy-radical overload diseases find increased levels of these enzymes (7188), others find a decrease (89100). Although these findings are contradicting, consideration should be given to the timing of the assay. Time-course studies can show an early increase in enzymatic activity/levels followed by a decrease (101). More importantly, consistent with the imbalance theme proposed in the above sections, the overall effect of the antioxidant system depends on the intracellular balance between these antioxidant enzymes rather than a single component. For example, increases in SOD in the absence of concomitant catalase and GPx increases would generate an excess amount of hydrogen peroxide and hydroxyl radicals (Figure 1). An imbalance has been shown to occur in tumor cells (102), in tissues undergoing chronic inflammation (91) and in smokers (103106). Recent studies indicate that p53 may play a role in this process (16).



View larger version (22K):
[in this window]
[in a new window]
 
Fig. 1. Free radical generation and protection from free radicals by SOD, catalase and GPx.

 
One molecule that could be categorized as a Jeckyl and Hyde antioxidant is nitric oxide (NO), first described as endothelium-derived relaxation factor (EDRF) in the 1980 s. NO is a key signaling molecule that mediates many physiological processes, including vasodilation, neurotransmission, host-defence, platelet aggregation and iron metabolism. There is conflicting evidence, however, that this molecule is anti- or pro-carcinogenic. Toward this end, NO has been shown to be an antioxidant (107) with a pro-carcinogenic activity, depending on the genetic background and surrounding tissue milieu (108). Further studies are therefore required to properly assess whether targeting NO for depletion will inhibit or contribute to carcinogenesis. Toward this end, the observation that inhibition of cyclooxygenases (1 and 2) exacerbates colitis and possibly colon cancer associated with this condition offers insight into the contradicting affects of these molecules on carcinogenesis (109111).


    Carcinogen metabolizing adaptation
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
A careful co-ordination and balance of phase I and phase II metabolizing enzymes is necessary for cellular protection against xenobiotics. These enzymes are present in abundance either at the basal level or are induced by genotoxic stress (112114), and are geared toward changing a bio-active compound into an inactive and excretable compound. Paradoxically, however, most of the intermediates following phase I metabolism are highly reactive with biological molecules, including DNA, RNA and proteins. Therefore, during xenobiotic metabolism, the balance between phase I and phase II enzymes can dramatically alter pharmacokinetics and xenobiotic toxicity. For example, a shift toward increasing phase I enzymatic activity in the absence of phase II changes, either through genetic polymorphisms or genotoxic stress, can exacerbate the impact of carcinogens (114).


    Conclusions and perspectives
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 
Complex pathways have evolved in mammalian cells aimed at maintaining cellular health. There are many ways to interpret why the expression of specific genes are changed in an environment of genotoxic stress, such as that in chronic inflammation. A very complex profile arises in chronically inflamed tissues, with the induction of pro- and anti-inflammatory cytokines, enzymes such as iNOS and cox-2, and proteins initially believed to protect our genome. Although this protective reaction to the insult sometimes has a beneficial effect, it is becoming increasingly clear that there is a snowball effect with feed-forward pro-mutagenic implications. Guo and Loeb (115) suggested ‘there are many ways that cells can tumble down the path toward genetic instability’. This is important to understand when assessing the use of these molecules in current cancer chemoprevention and treatment strategies. Information supplied by studies on genetic and functional polymorphisms will be beneficial in understanding the genetic components associated with how cells adapt to genotoxic stress.

An additional hypothesis that deserves attention is outlined in Figure 2. An accepted model is that of absolute cellular protection by adaptive increases in genome-protection pathways (Figure 2A). I have argued that this can have pro-carcinogenic consequences (Figure 2B). One model worth exploring, however, is that of intermittent genotoxic stress (Figure 2C). The genome-protection machinery adapts to a genotoxic environment. However, in the absence of this environment, this response is unnecessary, and the ‘protective’ machinery relaxes. Consequently, in a situation of sudden exposure to another or the same genotoxic stress, this machinery is not there. Therefore, there is more risk for genotoxic damage. If this periodic and sudden exposure occurs multiple times, this can have pro-carcinogenic consequences. An analogy of this exposure, with results consistent with this hypothesis, is the epidemiological finding of increased risk of skin cancer with multiple sunburns (116118). Another example of this would be individuals who quit and re-start smoking multiple times. The hypothesis is that the larger number of times a person quits smoking, the greater the increased lung cancer risk. In support of this hypothesis is the recent finding that cigarette smoke causes an adaptive increase in gene expression patterns, which return to normal when exposure is interrupted (106). However, it does not appear that this type of data has been reported in epidemiological studies, and further examination is necessary to explore such a hypothesis.



View larger version (44K):
[in this window]
[in a new window]
 
Fig. 2. Model depicting how chronic exposure to genotoxic stress (indicated by vertical arrows) leads to an adaptive increase in genome protecting proteins. An accepted understanding has been one of protection by the up-regulation of genome guardians (A). Genome guardian adaptive increases are represented by an increasing size of the cells. It is becoming increasingly clear that these genome guardians paradoxically have pro-mutagenic consequences (B). If this stress is not consistent, with intermittent lapses in genotoxic stress, there will be a down-regulation of these proteins. When the stress appears again, these proteins are not available for protection, possibly leading to a greater amount of cellular damage than with chronic exposure (C).

 

    Acknowledgments
 
Thanks to Dr Frank Berger (University of South Carolina), Dr Miriam Rosin (British Columbia Cancer Agency) and Dr David Christiani (Harvard) for their helpful comments. This work was supported in part by the COBRE funded Center for Colon Cancer Research, NIH Grant# P20 RR17698-01.


    References
 Top
 Abstract
 Introduction
 p53 adaptation
 DNA repair adaptation
 Cell cycle checkpoint adaptation
 Apoptosis adaptation
 Antioxidant adaptation
 Carcinogen metabolizing...
 Conclusions and perspectives
 References
 

  1. Harris,C.C. (1993) p53: at the crossroads of molecular carcinogenesis and risk assessment. Science, 262, 1980–1981.[ISI][Medline]
  2. Hofseth,L.J., Hussain S.P. and Harris C.C. (2004) p53: 25 years after its discovery. Trends Pharmacol. Sci., 25, 177–181.[CrossRef][ISI][Medline]
  3. Maltzman,W. and Czyzyk,L. (1984) UV irradiation stimulates levels of p53 cellular tumor antigen in nontransformed mouse cells. Mol. Cell. Biol., 4, 1689–1694.[ISI][Medline]
  4. Kastan,M.B., Onyekwere,O., Sidransky,D., Vogelstein,B. and Craig,R.W. (1991) Participation of p53 protein in the cellular response to DNA damage. Cancer Res., 51, 6304–6311.[Abstract]
  5. Kuerbitz,S.J., Plunkett,B.S., Walsh,W.V. and Kastan,M.B. (1992) Wild-type p53 is a cell cycle checkpoint determinant following irradiation. Proc. Natl Acad. Sci. USA, 89, 7491–7495.[Abstract]
  6. Hofseth,L.J., Saito,S., Hussain,S.P. et al. (2003) Nitric oxide-induced cellular stress and p53 activation in chronic inflammation. Proc. Natl Acad. Sci. USA, 100, 143–148.[Abstract/Free Full Text]
  7. Harvey,M., Vogel,H., Morris,D., Bradley,A., Bernstein,A. and Donehower,L.A. (1995) A mutant p53 transgene accelerates tumour development in heterozygous but not nullizygous p53-deficient mice. Nature Genet., 9, 305–311.[ISI][Medline]
  8. Zhang,Z., Liu,Q., Lantry,L.E., Wang,Y., Kelloff,G.J., Anderson,M.W., Wiseman,R.W., Lubet,R.A. and You,M. (2000) A germ-line p53 mutation accelerates pulmonary tumorigenesis: p53-independent efficacy of chemopreventive agents green tea or dexamethasone/myo-inositol and chemotherapeutic agents taxol or adriamycin. Cancer Res., 60, 901–907.[Abstract/Free Full Text]
  9. Wang,Y., Zhang,Z., Kastens,E., Lubet,R.A. and You,M. (2003) Mice with alterations in both p53 and Ink4a/Arf display a striking increase in lung tumor multiplicity and progression: differential chemopreventive effect of budesonide in wild-type and mutant A/J mice. Cancer Res., 63, 4389–4395.[Abstract/Free Full Text]
  10. De Flora,S., Balansky,R.M., D'Agostini,F., Izzotti,A., Camoirano,A., Bennicelli,C., Zhang,Z., Wang,Y., Lubet,R.A. and You,M. (2003) Molecular alterations and lung tumors in p53 mutant mice exposed to cigarette smoke. Cancer Res., 63, 793–800.[Abstract/Free Full Text]
  11. Duan,W., Ding,H., Subler,M.A., Zhu,W.G., Zhang,H., Stoner,G.D., Windle,J.J., Otterson,G.A. and Villalona-Calero,M.A. (2002) Lung-specific expression of human mutant p53-273H is associated with a high frequency of lung adenocarcinoma in transgenic mice. Oncogene, 21, 7831–7838.[CrossRef][ISI][Medline]
  12. Tyner,S.D., Venkatachalam,S., Choi,J. et al. (2002) p53 mutant mice that display early ageing-associated phenotypes. Nature, 415, 45–53.[CrossRef][ISI][Medline]
  13. Hussain,S.P., Amstad,P., Raja,K. et al. (2000) Increased p53 mutation load in noncancerous colon tissue from ulcerative colitis: a cancer-prone chronic inflammatory disease. Cancer Res., 60, 3333–3337.[Abstract/Free Full Text]
  14. Hussain,S.P., Raja,K., Amstad,P.A. et al. (2000) Increased p53 mutation load in nontumorous human liver of wilson disease and hemochromatosis: oxyradical overload diseases. Proc. Natl Acad. Sci. USA, 97, 12770–12775.[Abstract/Free Full Text]
  15. Polyak,K., Xia,Y., Zweier,J.L., Kinzler,K.W. and Vogelstein,B. (1997) A model for p53-induced apoptosis. Nature, 389, 300–305.[CrossRef][ISI][Medline]
  16. Hussain,S.P., Amstad,P., He,P. et al. (2004) p53-induced up-regulation of MnSOD and GPx but not catalase increases oxidative stress and apoptosis. Cancer Res., 64, 2350–2356.[Abstract/Free Full Text]
  17. Cerutti,P.A. (1985) Prooxidant states and tumor promotion. Science, 227, 375–381.[ISI][Medline]
  18. Weinstein,I.B. (2000) Disorders in cell circuitry during multistage carcinogenesis: the role of homeostasis. Carcinogenesis, 21, 857–864.[Abstract/Free Full Text]
  19. Weinberg,R.A. (1995) The retinoblastoma protein and cell cycle control. Cell, 81, 323–330.[ISI][Medline]
  20. Witkin,E.M. (1973) Ultraviolet mutagenesis in bacteria: the inducible nature of error-prone repair. An. Acad. Bras. Cienc., 45(suppl.), 185–191.[Medline]
  21. Radman,M. (1975) SOS repair hypothesis: phenomenology of an inducible DNA repair which is accompanied by mutagenesis. Basic Life Sci., 5A, 355–367.[Medline]
  22. Fritz,G., Tano,K., Mitra,S. and Kaina,B. (1991) Inducibility of the DNA repair gene encoding O6-methylguanine-DNA methyltransferase in mammalian cells by DNA-damaging treatments. Mol. Cell. Biol., 11, 4660–4668.[ISI][Medline]
  23. Fornace,A.J.,Jr, Zmudzka,B., Hollander,M.C. and Wilson,S.H. (1989) Induction of beta-polymerase mRNA by DNA-damaging agents in Chinese hamster ovary cells. Mol. Cell. Biol., 9, 851–853.[ISI][Medline]
  24. Laval,F. (1991) Related increase of O6-methylguanine-DNA-methyltransferase and N3-methyladenine glycosylase RNA transcripts in rat hepatoma cells treated with DNA-damaging agents. Biochem. Biophys. Res. Commun., 176, 1086–1092.[ISI][Medline]
  25. Ramana,C.V., Boldogh,I., Izumi,T. and Mitra,S. (1998) Activation of apurinic/apyrimidinic endonuclease in human cells by reactive oxygen species and its correlation with their adaptive response to genotoxicity of free radicals. Proc. Natl Acad. Sci. USA, 95, 5061–5066.[Abstract/Free Full Text]
  26. Lee,Y.S., Choi,J.Y., Park,M.K., Choi,E.M., Kasai,H. and Chung,M.H. (1996) Induction of oh8Gua glycosylase in rat kidneys by potassium bromate (KBrO3), a renal oxidative carcinogen. Mutat. Res., 364, 227–233.[ISI][Medline]
  27. Rusyn,I., Denissenko,M.F., Wong,V.A,, Butterworth,B.E., Cunningham,M.L., Upton,P.B., Thurman,R.G. and Swenberg,J.A. (2000) Expression of base excision repair enzymes in rat and mouse liver is induced by peroxisome proliferators and is dependent upon carcinogenic potency. Carcinogenesis, 21, 2141–2145[Abstract/Free Full Text]
  28. Rusyn,I., Asakura,S., Pachkowski,B., Bradford,B.U., Denissenko,M.F., Peters,J.M., Holland,S.M., Reddy,J.K., Cunningham,M.L. and Swenberg,J.A. (2003) Expression of base excision DNA repair genes is a sensitive biomarker for in vivo detection of chemical-induced chronic oxidative stress: identification of the molecular source of radicals responsible for DNA damage by peroxisome proliferators. Cancer Res., 64, 1050–1057.[ISI]
  29. Hofseth,L.J., Khan,M.A., Ambrose,M. et al. (2003) The adaptive imbalance in base excision-repair enzymes generates microsatellite instability in chronic inflammation. J. Clin. Invest., 112, 1887–1894.[Abstract/Free Full Text]
  30. Xiao,W. and Samson,L. (1993) In vivo evidence for endogenous DNA alkylation damage as a source of spontaneous mutation in eukaryotic cells. Proc. Natl Acad. Sci. USA, 90, 2117–2121.[Abstract]
  31. Shcherbakova,P.V. and Kunkel,T.A. (1999) Mutator phenotypes conferred by MLH1 overexpression and by heterozygosity for mlh1 mutations. Mol. Cell. Biol., 19, 3177–3183.[Abstract/Free Full Text]
  32. Shcherbakova,P.V., Hall,M.C., Lewis,M.S., Bennett,S.E., Martin,K.J., Bushel,P.R., Afshari,C.A. and Kunkel,T.A. (2001) Inactivation of DNA mismatch repair by increased expression of yeast MLH1. Mol. Cell. Biol., 21, 940–951.[Abstract/Free Full Text]
  33. Bergoglio,V., Pillaire,M.J., Lacroix-Triki,M. et al. (2002) Deregulated DNA polymerase beta induces chromosome instability and tumorigenesis. Cancer Res., 62, 3511–3514.[Abstract/Free Full Text]
  34. Yamada,N.A. and Farber,R.A. (2002) Induction of a low level of microsatellite instability by overexpression of DNA polymerase Beta. Cancer Res., 62, 6061–6064.[Abstract/Free Full Text]
  35. Weinert,T.A. and Hartwell,L.H. (1988) The RAD9 gene controls the cell cycle response to DNA damage in Saccharomyces cerevisiae. Science, 241, 317–322.[ISI][Medline]
  36. Harper,J.W., Adami,G.R., Wei,N., Keyomarsi,K. and Elledge,S.J. (1993) The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell, 75, 805–816.[ISI][Medline]
  37. Bunz,F., Dutriaux,A., Lengauer,C., Waldman,T., Zhou,S., Brown,J.P., Sedivy,J.M., Kinzler,K.W. and Vogelstein,B. (1998) Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science, 282, 1497–1501.[Abstract/Free Full Text]
  38. Wang,X.W., Zhan,Q., Coursen,J.D., Khan,M.A., Kontny,H.U., Yu,L., Hollander,M.C., O'Connor,P.M., Fornace,A.J.,Jr and Harris,C.C. (1999) GADD45 induction of a G2/M cell cycle checkpoint. Proc. Natl Acad. Sci. USA, 96, 3706–3711.[Abstract/Free Full Text]
  39. Liu,J., Kadiiska,M.B., Liu,Y., Lu,T., Qu,W. and Waalkes,M.P. (2001) Stress-related gene expression in mice treated with inorganic arsenicals. Toxicol. Sci., 61, 314–320.[Abstract/Free Full Text]
  40. Lu,Y.P., Lou,Y.R., Yen,P., Mitchell,D., Huang,M.T. and Conney,A.H. (1999) Time course for early adaptive responses to ultraviolet B light in the epidermis of SKH-1 mice. Cancer Res., 59, 591–602.
  41. Smith,M.L., Kontny,H.U., Zhan,Q., Sreenath,A., O'Connor,P.M. and Fornace,A.J.,Jr (1996) Antisense GADD45 expression results in decreased DNA repair and sensitizes cells to u.v.-irradiation or cisplatin. Oncogene, 13, 2255–2263.[ISI][Medline]
  42. Smith,M.L., Ford,J.M., Hollander,M.C., Bortnick,R.A., Amundson,S.A., Seo,Y.R., Deng,C.X., Hanawalt,P.C. and Fornace,A.J.,Jr (2000) p53-mediated DNA repair responses to UV radiation: studies of mouse cells lacking p53, p21 and/or gadd45 genes. Mol. Cell. Biol., 20, 3705–3714.[Abstract/Free Full Text]
  43. Roninson,I.B. (2002) Oncogenic functions of tumour suppressor p21 (Waf1/Cip1/Sdi1): association with cell senescence and tumour-promoting activities of stromal fibroblasts. Cancer Lett., 179, 1–14.[CrossRef][ISI][Medline]
  44. Wang,Y.A., Elson,A. and Leder,P. (1997) Loss of p21 increases sensitivity to ionizing radiation and delays the onset of lymphoma in atm-deficient mice. Proc. Natl Acad. Sci. USA, 94, 14590–14595.[Abstract/Free Full Text]
  45. Weiss,R.H., Joo,A. and Randour,C. (2000) p21 (Waf1/Cip1) is an assembly factor required for platelet-derived growth factor-induced vascular smooth muscle cell proliferation. J. Biol. Chem., 275, 10285–10290.[Abstract/Free Full Text]
  46. Dong,Y., Chi,S.L., Borowsky,A.D., Fan,Y. and Weiss,R.H. (2004) Cytosolic p21Waf1/Cip1 increases cell cycle transit in vascular smooth muscle cells. Cell Signal., 16, 263–269.[CrossRef][ISI][Medline]
  47. Komiya,T., Hosono,Y., Hirashima,T., Masuda,N., Yasumitsu,T., Nakagawa,K., Kikui,M., Ohno,A., Fukuoka,M. and Kawase,I. (1997) p21 expression as a predictor for favorable prognosis in squamous cell carcinoma of the lung. Clin. Cancer Res., 3, 1831–1835.[Abstract]
  48. Zirbes,T.K., Baldus,S.E., Moenig,S.P. et al. (2000) Prognostic impact of p21/waf1/cip1 in colorectal cancer. Int. J. Cancer, 89, 14–18.[CrossRef][ISI][Medline]
  49. Lu,X., Toki,T., Konishi,I., Nikaido,T. and Fujii,S. (1998) Expression of p21WAF1/CIP1 in adenocarcinoma of the uterine cervix: a possible immunohistochemical marker of a favorable prognosis. Cancer, 82, 2409–2417.[CrossRef][ISI][Medline]
  50. Kapranos,N., Stathopoulos,G.P., Manolopoulos,L., Kokka,E., Papadimitriou,C., Bibas,A., Yiotakis,J. and Adamopoulos,G. (2001) p53, p21 and p27 protein expression in head and neck cancer and their prognostic value. Anticancer Res., 21, 521–528.[ISI][Medline]
  51. Roman-Gomez,J., Castillejo,J.A., Jimenez,A., Gonzalez,M.G., Moreno,F., Rodriguez,M.C., Barrios,M., Maldonado,J. and Torres,A. (2002) 5' CpG island hypermethylation is associated with transcriptional silencing of the p21 (CIP1/WAF1/SDI1) gene and confers poor prognosis in acute lymphoblastic leukemia. Blood, 99, 2291–2296.[Abstract/Free Full Text]
  52. Baldus,S.E., Schneider,P.M., Monig,S.P. et al. (2001) p21/waf1/cip1 in gastric cancer: associations with histopathological subtypes, lymphonodal metastasis, prognosis and p53 status. Scand. J. Gastroenterol., 36, 975–980.[ISI][Medline]
  53. Baretton,G.B., Klenk,U., Diebold,J., Schmeller,N. and Lohrs,U. (1999) Proliferation- and apoptosis-associated factors in advanced prostatic carcinomas before and after androgen deprivation therapy: prognostic significance of p21/WAF1/CIP1 expression. Br. J. Cancer, 80, 546–555.[CrossRef][ISI][Medline]
  54. Aaltomaa,S., Lipponen,P., Eskelinen,M., Ala-Opas,M. and Kosma,V.M. (1999) Prognostic value and expression of p21 (waf1/cip1) protein in prostate cancer. Prostate, 39, 8–15.[CrossRef][ISI][Medline]
  55. Ferrandina,G., Stoler,A., Fagotti,A., Fanfani,F., Sacco,R., De Pasqua,A., Mancuso,S. and Scambia,G. (2000) p21WAF1/CIP1 protein expression in primary ovarian cancer. Int. J. Oncol., 17, 1231–11235.[ISI][Medline]
  56. Cheung,T.H., Lo,K.W., Yu,M.M., Yim,S.F., Poon,C.S., Chung,T.K. and Wong,Y.F. (2001) Aberrant expression of p21 (WAF1/CIP1) and p27 (KIP1) in cervical carcinoma. Cancer Lett., 172, 93–98.[CrossRef][ISI][Medline]
  57. Bae,D.S., Cho,S.B., Kim,Y.J., Whang,J.D., Song,S.Y., Park,C.S., Kim,D.S. and Lee,J.H. (2001) Aberrant expression of cyclin D1 is associated with poor prognosis in early stage cervical cancer of the uterus. Gynecol. Oncol., 81, 341–347.[CrossRef][ISI][Medline]
  58. Omar,E.A., Behlouli,H., Chevalier,S. and Aprikian,A.G. (2001) Relationship of p21 (WAF-I) protein expression with prognosis in advanced prostate cancer treated by androgen ablation. Prostate, 49, 191–199.[CrossRef][ISI][Medline]
  59. Niculescu,A.B. 3rd, Chen,X., Smeets,M., Hengst,L., Prives,C. and Reed,S.I. (1998) Effects of p21 (Cip1/Waf1) at both the G1/S and the G2/M cell cycle transitions: pRb is a critical determinant in blocking DNA replication and in preventing endoreduplication. Mol. Cell. Biol., 18, 629–643.[Abstract/Free Full Text]
  60. Bates,S., Ryan,K.M., Phillips,A.C. and Vousden,K.H. (1998) Cell cycle arrest and DNA endoreduplication following p21Waf1/Cip1 expression. Oncogene, 17, 1691–1703.[CrossRef][ISI][Medline]
  61. Chang,B.D., Broude,E.V., Fang,J., Kalinichenko,T.V., Abdryashitov,R., Poole,J.C. and Roninson,I.B. (2000) p21Waf1/Cip1/Sdi1-induced growth arrest is associated with depletion of mitosis-control proteins and leads to abnormal mitosis and endoreduplication in recovering cells. Oncogene, 19, 2165–2170.[CrossRef][ISI][Medline]
  62. Chang,B.D, Watanabe,K., Broude,E.V., Fang,J., Poole,J.C., Kalinichenko,T.V. and Roninson,I.B. (2000) Effects of p21Waf1/Cip1/Sdi1 on cellular gene expression: implications for carcinogenesis, senescence and age-related diseases. Proc. Natl Acad. Sci. USA, 97, 4291–4296.[Abstract/Free Full Text]
  63. Danial,N.N. and Korsmeyer,S.J. (2004) Cell death: critical control points. Cell, 116, 205–219.[CrossRef][ISI][Medline]
  64. Jungas,T., Motta,I., Duffieux,F., Fanen,P., Stoven,V. and Ojcius,D.M. (2002) Glutathione levels and BAX activation during apoptosis due to oxidative stress in cells expressing wild-type and mutant cystic fibrosis transmembrane conductance regulator. J. Biol. Chem., 277, 27912–27918.[Abstract/Free Full Text]
  65. Ramp,U., Caliskan,E., Mahotka,C., Krieg,A., Heikaus,S., Gabbert,H.E. and Gerharz,C.D. (2003) Apoptosis induction in renal cell carcinoma by TRAIL and gamma-radiation is impaired by deficient caspase-9 cleavage. Br. J. Cancer, 88, 1800–1807.[CrossRef][ISI][Medline]
  66. Dunkern,T.R., Fritz,G. and Kaina,B. (2001) Ultraviolet light-induced DNA damage triggers apoptosis in nucleotide excision repair-deficient cells via Bcl-2 decline and caspase-3/-8 activation. Oncogene, 20, 6026–6038.[CrossRef][ISI][Medline]
  67. Zhan,Q., Fan,S., Bae,I., Guillouf,C., Liebermann,D.A., O'Connor,P.M. and Fornace,A.J.,Jr (1994) Induction of bax by genotoxic stress in human cells correlates with normal p53 status and apoptosis. Oncogene, 9, 3743–3751.[ISI][Medline]
  68. Wu,G.S., Kim,K. and el-Deiry,W.S. (2000) KILLER/DR5, a novel DNA-damage inducible death receptor gene, links the p53-tumor suppressor to caspase activation and apoptotic death. Adv. Exp. Med. Biol., 465, 143–151.[ISI][Medline]
  69. Suhara,T., Fukuo,K., Sugimoto,T., Morimoto,S., Nakahashi,T., Hata,S., Shimizu,M. and Ogihara,T. (1998) Hydrogen peroxide induces up-regulation of Fas in human endothelial cells. J. Immunol., 160, 4042–4047.[Abstract/Free Full Text]
  70. Cerutti,P., Larsson,R., Krupitza,G., Muehlematter,D., Crawford,D. and Amstad,P. (1988) Pathophysiological mechanisms of oxidants. In Cerutti PF (ed.) Oxy-Radicals in Molecular Biology and Pathology. Alan R. Liss, New York.
  71. Dong,H., Toyoda,N., Yoneyama,H., Kurachi,M., Kasahara,T., Kobayashi,Y., Inadera,H., Hashimoto,S. and Matsushima,K. (2002) Gene expression profile analysis of the mouse liver during bacteria-induced fulminant hepatitis by a cDNA microarray system. Biochem. Biophys. Res. Commun., 298, 675–686.[CrossRef][ISI][Medline]
  72. Chiu,H., Brittingham,J.A. and Laskin,D.L. (2002) Differential induction of heme oxygenase-1 in macrophages and hepatocytes during acetaminophen-induced hepatotoxicity in the rat: effects of hemin and biliverdin. Toxicol. Appl. Pharmacol., 181, 106–115.[CrossRef][ISI][Medline]
  73. Bauer,I., Vollmar,B., Jaeschke,H., Rensing,H., Kraemer,T., Larsen,R. and Bauer,M. (2000) Transcriptional activation of heme oxygenase-1 and its functional significance in acetaminophen-induced hepatitis and hepatocellular injury in the rat. J. Hepatol., 33, 395–406.[CrossRef][ISI][Medline]
  74. Wang,W.P., Guo,X., Koo,M.W., Wong,B.C., Lam,S.K., Ye,Y.N. and Cho,C.H. (2001) Protective role of heme oxygenase-1 on trinitrobenzene sulfonic acid-induced colitis in rats. Am. J. Physiol. Gastrointest. Liver Physiol., 281, G586–G594.[Abstract/Free Full Text]
  75. Fu,K., Tomita,T., Sarras,M.P.,Jr, De Lisle,R.C. and Andrews,G.K. (1998) Metallothionein protects against cerulein-induced acute pancreatitis: analysis using transgenic mice. Pancreas, 17, 238–246.[ISI][Medline]
  76. Fu,K., Sarras,M.P.,Jr, De Lisle,R.C. and Andrews,G.K. (1997) Expression of oxidative stress-responsive genes and cytokine genes during caerulein-induced acute pancreatitis. Am. J. Physiol., 273, G696–705.[ISI][Medline]
  77. Barbosa,D.S., Cecchini,R., El Kadri,M.Z., Rodriguez,M.A., Burini,R.C. and Dichi,I. (2003) Decreased oxidative stress in patients with ulcerative colitis supplemented with fish oil omega-3 fatty acids. Nutrition, 19, 837–842.[CrossRef][ISI][Medline]
  78. Ding,S.P., Li,J.C. and Jin,C. (2003) A mouse model of severe acute pancreatitis induced with caerulein and lipopolysaccharide. World J. Gastroenterol., 9, 584–589.[ISI][Medline]
  79. Su,S.B., Motoo,Y., Xie,M.J., Mouri,H., Asayama,K. and Sawabu,N. (2002) Superoxide dismutase is induced during rat pancreatic acinar cell injury. Pancreas, 24, 146–152.[CrossRef][ISI][Medline]
  80. Abe,R., Shimosegawa,T., Moriizumi,S., Kikuchi,Y., Kimura,K., Satoh,A., Koizumi,M. and Toyota,T. (1995) Lipopolysaccharide induces manganese superoxide dismutase in the rat pancreas: its role in caerulein pancreatitis. Biochem. Biophys. Res. Commun., 217, 1216–1222.[CrossRef][ISI][Medline]
  81. Sihvo,E.I., Salminen,J.T., Rantanen,T.K., Ramo,O.J., Ahotupa,M., Farkkila,M., Auvinen,M.I. and Salo,J.A. (2002) Oxidative stress has a role in malignant transformation in Barrett's oesophagus. Int. J. Cancer, 102, 551–555.[CrossRef][ISI][Medline]
  82. Szuster-Ciesielska,A., Daniluk,J. and Kandefer-Szerszen,M. (2001) Oxidative stress in blood of patients with alcohol-related pancreatitis. Pancreas, 22, 261–266.[CrossRef][ISI][Medline]
  83. Mathew,P., Wyllie,R., Van Lente,F., Steffen,R.M. and Kay,M.H. (1996) Antioxidants in hereditary pancreatitis. Am. J. Gastroenterol., 91, 1558–1562.[ISI][Medline]
  84. Toborek,M., Kopieczna-Grzebieniak,E., Drozdz,M. and Wieczorek,M. (1996) Increased lipid peroxidation and antioxidant activity in methionine-induced hepatitis in rabbits. Nutrition, 12, 534–537.[CrossRef][ISI][Medline]
  85. Valentine,J.F., Tannahill,C.L., Stevenot,S.A., Sallustio,J.E., Nick,H.S. and Eaker,E.Y. (1996) Colitis and interleukin 1beta up-regulate inducible nitric oxide synthase and superoxide dismutase in rat myenteric neurons. Gastroenterology, 111, 56–64.[ISI][Medline]
  86. Valentine,J.F. and Nick,H.S. (1994) Glucocorticoids repress basal and stimulated manganese superoxide dismutase levels in rat intestinal epithelial cells. Gastroenterology, 107, 1662–1670.[ISI][Medline]
  87. Demirdag,K., Yilmaz,S., Ozdarendeli,A., Ozden,M., Kalkan,A. and Kilic,S.S. (2003) Levels of plasma malondialdehyde and erythrocyte antioxidant enzyme activities in patients with chronic hepatitis B. Hepatogastroenterology, 50, 766–770.[ISI][Medline]
  88. Sanchez-Bernal,C., Garcia-Morales,O.H., Dominguez,C., Martin-Gallan,P., Calvo,J.J., Ferreira,L. and Perez-Gonzalez,N. (2003) Nitric oxide protects against pancreatic subcellular damage in acute pancreatitis. Pancreas, 28, e9–15.[ISI]
  89. Dong,W.G., Mei,Q., Yu,J.P., Xu,J.M., Xiang,L. and Xu,Y. (2003) Effects of melatonin on the expression of iNOS and COX-2 in rat models of colitis. World J. Gastroenterol, 9, 1307–1311.[ISI][Medline]
  90. Park,B.K., Chung,J.B., Lee,J.H., Suh,J.H., Park S.W., Song,S.Y., Kim,H., Kim,K.H. and Kang,J.K. (2003) Role of oxygen free radicals in patients with acute pancreatitis. World J. Gastroenterol., 9, 2266–2269.[ISI][Medline]
  91. Kruidenier,L., Kuiper,I., van Duijn,W., Marklund,S.L., van Hogezand,R.A., Lamers,C.B. and Verspaget,H.W. (2003) Differential mucosal expression of three superoxide dismutase isoforms in inflammatory bowel disease. J. Pathol., 201, 7–16.[CrossRef][ISI][Medline]
  92. Cullen,J.J., Mitros,F.A. and Oberley,L.W. (2003) Expression of antioxidant enzymes in diseases of the human pancreas: another link between chronic pancreatitis and pancreatic cancer. Pancreas, 26, 23–27.[CrossRef][ISI][Medline]
  93. Zeki,S., Miura,S., Suzuki,H., Watanabe,N., Adachi,M., Yokoyama,H., Horie,Y., Saito,H., Kato,S. and Ishii,H. (2002) Xanthine oxidase-derived oxygen radicals play significant roles in the development of chronic pancreatitis in WBN/Kob rats. J. Gastroenterol. Hepatol., 17, 606–616.[CrossRef][ISI][Medline]
  94. Korenaga,D., Takesue,F., Kido,K., Yasuda,M., Inutsuka,S., Honda,M. and Nagahama,S. (2002) Impaired antioxidant defense system of colonic tissue and cancer development in dextran sulfate sodium-induced colitis in mice. J. Surg. Res., 102, 144–149.[CrossRef][ISI][Medline]
  95. Al-Awadi,F.M., Srikumar,T.S., Anim,J.T. and Khan,I. (2001) Antiinflammatory effects of Cordia myxa fruit on experimentally induced colitis in rats. Nutrition, 17, 391–396.[CrossRef][ISI][Medline]
  96. Czako,L., Takacs,T., Varga,I.S., Tiszlavicz,L., Hai,D.Q., Hegyi,P., Matkovics,B. and Lonovics,J. (1998) Involvement of oxygen-derived free radicals in L-arginine-induced acute pancreatitis. Dig. Dis. Sci., 43, 1770–1777.[CrossRef][ISI][Medline]
  97. Lih-Brody,L., Powell,S.R., Collier,K.P. et al. (1996) Increased oxidative stress and decreased antioxidant defenses in mucosa of inflammatory bowel disease. Dig. Dis. Sci., 41, 2078–2086.[ISI][Medline]
  98. Seo,H.G., Takata,I., Nakamura,M., Tatsumi,H., Suzuki,K., Fujii,J. and Taniguchi,N. (1995) Induction of nitric oxide synthase and concomitant suppression of superoxide dismutases in experimental colitis in rats. Arch. Biochem. Biophys., 324, 41–47.[CrossRef][ISI][Medline]
  99. Wetscher,G.J., Perdikis,G., Kretchmar,D.H., Stinson,R.G., Bagchi,D., Redmond,E.J., Adrian,T.E. and Hinder,R.A. (1995) Esophagitis in Sprague–Dawley rats is mediated by free radicals. Dig. Dis. Sci., 40, 1297–1305.[ISI][Medline]
  100. Nonaka,A., Manabe,T., Tamura,K., Asano,N., Imanishi,K. and Tobe,T. (1989) Changes of xanthine oxidase, lipid peroxide and superoxide dismutase in mouse acute pancreatitis. Digestion, 43, 41–56.[ISI][Medline]
  101. Loguercio,C., D'Argenio,G., Delle Cave,M., Cosenza,V., Della Valle,N., Mazzacca,G. and del Vecchio Blanco,C. (1996) Direct evidence of oxidative damage in acute and chronic phases of experimental colitis in rats. Dig. Dis. Sci., 41, 1204–1211.[ISI][Medline]
  102. Policastro,L., Molinari,B., Larcher,F., Blanco,P., Podhajcer,O.L., Costa,C.S., Rojas,P. and Duran,H. (2004) Imbalance of antioxidant enzymes in tumor cells and inhibition of proliferation and malignant features by scavenging hydrogen peroxide. Mol. Carcinogen., 39, 103–113.[CrossRef][ISI][Medline]
  103. Yildiz,L., Kayaoglu,N. and Aksoy,H. (2002) The changes of superoxide dismutase, catalase and glutathione peroxidase activities in erythrocytes of active and passive smokers. Clin. Chem. Lab. Med., 40, 612–615.[ISI][Medline]
  104. Rajpurkar,A., Dhabuwala C.B., Jiang,Y. and Li,H. (2000) Chronic cigarette smoking induces an oxidant–antioxidant imbalance in the testis. J. Environ. Pathol. Toxicol. Oncol., 19, 369–373.[Medline]
  105. MacNee,W. (2000) Oxidants/antioxidants and COPD. Chest, 117, 303S–317S.[Free Full Text]
  106. Gebel,S., Gerstmayer,B., Bosio,A., Haussmann,H.J., Van Miert,E. and Muller,T. (2004) Gene expression profiling in respiratory tissues from rats exposed to mainstream cigarette smoke. Carcinogenesis, 25, 169–178.[Abstract/Free Full Text]
  107. Wink,D.A., Hanbauer,I., Krishna,M.C., DeGraff,W., Gamson,J. and Mitchell,J.B. (1993) Nitric oxide protects against cellular damage and cytotoxicity from reactive oxygen species. Proc. Natl Acad. Sci. USA, 90, 9813–9817.[Abstract]
  108. Hofseth,L.J., Hussain,S.P., Wogan,G.N. and Harris,C.C. (2003) Nitric oxide in cancer and chemoprevention. Free. Radic. Biol. Med., 34, 955–968.[CrossRef][ISI][Medline]
  109. Reuter,B.K., Asfaha,S., Buret,A., Sharkey,K.A. and Wallace,J.L. (1996) Exacerbation of inflammation-associated colonic injury in rat through inhibition of cyclooxygenase-2. J. Clin. Invest., 98, 2076–2085.[Abstract/Free Full Text]
  110. Morteau,O., Morham,S.G., Sellon,R., Dieleman,L.A., Langenbach,R., Smithies,O. and Sartor,R.B. (2000) Impaired mucosal defense to acute colonic injury in mice lacking cyclooxygenase-1 or cyclooxygenase-2. J. Clin. Invest., 105, 469–478.[Abstract/Free Full Text]
  111. Hegazi,R.A., Mady,H.H., Melhem,M.F., Sepulveda,A.R., Mohi,M. and Kandil,H.M. (2003) Celecoxib and rofecoxib potentiate chronic colitis and premalignant changes in interleukin 10 knockout mice. Inflamm. Bowel Dis., 9, 230–236.[ISI][Medline]
  112. Hatanaka,N., Yamazaki,H., Kizu,R., Hayakawa,K., Aoki,Y., Iwanari,M., Nakajima,M. and Yokoi,T. (2001) Induction of cytochrome P450 1B1 in lung, liver and kidney of rats exposed to diesel exhaust. Carcinogenesis, 22, 2033–2038.[Abstract/Free Full Text]
  113. Villard,P.H., Seree,E.M., Re,J.L., De Meo,M., Barra,Y., Attolini,L., Dumenil,G., Catalin,J., Durand,A. and Lacarelle,B. (1998) Effects of tobacco smoke on the gene expression of the Cyp1a, Cyp2b, Cyp2e and Cyp3a subfamilies in mouse liver and lung: relation to single strand breaks of DNA. Toxicol. Appl. Pharmacol., 148, 195–204.[CrossRef][ISI][Medline]
  114. Ingelman-Sundberg,M. (2004) Pharmacogenetics of cytochrome P450 and its applications in drug therapy: the past, present and future. Trends Pharmacol. Sci., 25, 193–200.[CrossRef][ISI][Medline]
  115. Guo,H.H. and Loeb,L.A. (2003) Tumbling down a different pathway to genetic instability. J. Clin. Invest., 112, 1793–1795.[Abstract/Free Full Text]
  116. Veierod,M.B., Weiderpass,E., Thorn,M., Hansson,J., Lund,E., Armstrong,B. and Adami,H.O. (2003) A prospective study of pigmentation, sun exposure and risk of cutaneous malignant melanoma in women. J. Natl Cancer Inst., 95, 1530–1538.[Abstract/Free Full Text]
  117. Green,A., Siskind,V., Bain,C. and Alexander,J. (1985) Sunburn and malignant melanoma. Br. J. Cancer, 51, 393–397.[ISI][Medline]
  118. Harrison,S.L., MacLennan,R., Speare,R. and Wronski,I. (1994) Sun exposure and melanocytic naevi in young Australian children. Lancet, 344, 1529–1532.[ISI][Medline]
  119. Brancho,D., Tanaka,N., Jaeschke,A., Ventura,J.J., Kelkar,N., Tanaka,Y., Kyuuma,M., Takeshita,T., Flavell,R.A. and Davis,R.J. (2003) Mechanism of p38 MAP kinase activation in vivo. Genes Dev., 17, 1969–1978.[Abstract/Free Full Text]
  120. Bulavin,D.V., Amundson,S.A. and Fornace,A.J. (2002) p38 and Chk1 kinases: different conductors for the G(2)/M checkpoint symphony. Curr. Opin. Genet. Dev., 12, 92–97.[CrossRef][ISI][Medline]
  121. Abraham,R.T. (2001) Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev., 15, 2177–2196.[Free Full Text]
  122. Hirao,A., Kong,Y.Y., Matsuoka,S., Wakeham,A., Ruland,J., Yoshida,H., Liu,D., Elledge,S.J. and Mak,T.W. (2000) DNA damage-induced activation of p53 by the checkpoint kinase Chk2. Science, 287, 1824–1827.[Abstract/Free Full Text]
Received March 10, 2004; revised April 22, 2004; accepted May 18, 2004.