Induction of cyclin-dependent kinase inhibitors and G1 prolongation by the chemopreventive agent N-acetylcysteine
Ming Liu1,
Norbert M. Wikonkal1 and
Douglas E. Brash1,2,3
1 Department of Therapeutic Radiology and
2 Department of Genetics, Yale University School of Medicine, New Haven, CT 06520-8040, USA
 |
Abstract
|
---|
Cyclin-dependent kinase (cdk) inhibitors, such as p16INK4a and p21WAF1/CIP1, often inhibit G1 cyclin kinases and result in G1 arrest. It has been suggested that p21WAF1/CIP1 may also play a role in other chemopreventive activities such as DNA repair, slowdown of DNA replication and induction of cellular differentiation. In this report we demonstrate that the antioxidant N-acetylcysteine (NAC), a well-known chemopreventive agent, induces p16INK4a and p21WAF1/CIP1 gene expression and prolongs cell-cycle transition through G1 phase. A portion of the G1 arrest by NAC is governed by p16INK4a; it is independent of p53. NAC's usual mechanism of increasing intracellular glutathione level is not required for the G1 arrest. An antioxidant whose action is limited to scavenging radicals, Trolox, does not induce G1 arrest. Taken together, these results suggest a potential novel molecular basis for chemoprevention by NAC.
Abbreviations: BSO, buthionine sulfoximine; cdk, cyclin-dependent kinase; GSH, glutathione; NAC, N-acetylcysteine.
 |
Introduction
|
---|
N-acetylcysteine (NAC) is known as a chemopreventive agent with anti-mutagenic and anti-tumorigenic activities in a variety of animal organs, including skin, lung, liver and colon (16). As an antioxidant, NAC can act as a scavenger of reactive oxidative intermediates either by itself or indirectly. In the latter role, it serves as a precursor for glutathione (GSH;
-glutamylcysteinylglycine), the major intracellular thiol and antioxidant (79). NAC has been shown to block both DNA strand breakage and mutagenesis by DNA-damaging agents (1012). NAC is a small molecule readily taken up by cells and is used in clinical treatments for chronic bronchitis and paracetamol poisoning (13).
In a previous study, we reported that NAC induces p53-dependent apoptosis selectively in oncogenically-transformed cells (14). In contrast, chain-breaking antioxidants such as vitamin E and Trolox lacked this activity. An increased GSH level was not required for apoptosis by NAC. Agents that induce apoptosis often cause cell-cycle arrest as well, typically at lower doses. Here, we investigate the effect of NAC on cell cycle-progression and on expression of two cyclin-dependent kinase (cdk) inhibitors. We find that NAC elicits p16INK4a-mediated G1-phase prolongation and upregulates p21WAF1/CIP1 expression through a p53-independent pathway. These findings may have implications for the chemopreventive activities of NAC.
 |
Materials and methods
|
---|
Cell culture
The 308 papilloma cell line (wild-type p53) (15), the p53-null mouse keratinocyte cell line NHK-4 (16), and the mouse embryo fibroblast cell line (10)1 (17) were kindly provided by Drs S.Yuspa, W.Weinberg and A.Levine (National Cancer Institute, Rockefeller University), respectively. Human primary foreskin fibroblasts were supplied by the core facility of the Department of Dermatology, Yale School of Medicine, New Haven, CT. BALB/c 3T3 A31 cells were obtained from the American Type Culture Collection (Mannassas, VA). 308 papilloma cells were maintained in 0.05 mM Ca2+ Eagle's minimum essential medium with 10% fetal bovine serum, and all other cells were grown in Dulbecco's modified Eagle's medium with 10% heat-inactivated fetal bovine serum. Primary keratinocytes from newborn mouse skin were prepared as described previously (18). p21WAF1/CIP1 and INK4a knockout mice (19,20) were generously provided by Drs P.Leder (Harvard University, Boston, MA) and M.Serrano (Centro Nazional de Biotecnologia, Madrid), respectively.
Chemicals and cell treatments
NAC and Trolox were purchased from Sigma (St Louis, MO) and Aldrich (Milwaukee, WI), respectively. These were freshly dissolved in culture medium and adjusted to neutral pH if necessary. Culture confluence was maintained below 80%.
Northern and western blot analysis
Northern and western blot analyses were performed as described previously (21). In the analysis of p21WAF1/CIP1 and p16INK4a protein, rabbit p21WAF1/CIP1polyclonal antibody sc-397, mouse p16INK4a monoclonal antibody sc-1661 (Santa Cruz Biotechnology, Santa Cruz, CA), goat anti-rabbit IgG peroxidase conjugate (ICN Biomedicals, Costa Mesa, CA) and rabbit anti-mouse IgG peroxidase conjugate (Santa Cruz Biotechnology) were used as primary and secondary antibodies.
Flow cytometry
For each sample, ~106 cells were washed with ice-cold PBS and fixed in 95% ethanol. Cells were then resuspended in 1 mg/ml RNase (Sigma) for 30 min at 37°C and stained with 0.05 mg/ml propidium iodide (Sigma) for 1 h on ice. Flow cytometric analysis was performed with a FACS Vantage flow cytometer (Becton Dickinson, Franklin Lakes, NJ). Cells were excited at 488 nm and the emission was detected through a 630 ± 11 nm band pass filter. A minimum of 10 000 cells were analyzed for each sample. Cell-cycle analysis was performed using Modfit 5.2 software (Verity Software House, Topsham, ME).
 |
Results
|
---|
We first investigated the cell-cycle distribution of NAC-treated 308 cells. As shown in Table I
, the cells accumulated in G0/G1 phase within 48 h, with an accompanying decrease of cells in both S phase and G2/M phase. This G1 cell-cycle arrest was significant at an NAC dose of 5 mM. Lower levels of G0/G1 accumulation could be observed at a dose of 1 mM. This observation was reproduced in the BALB/c 3T3 A31 fibroblast cell line (data not shown), indicating that this arrest by NAC is not cell-type specific.
Cell-cycle progression is precisely regulated by a series of cyclins, cdks and cdk inhibitors. Given the known functions of cdk inhibitors p21WAF1/CIP1 and p16INK4a in controlling cell-cycle progression at G1, we next examined the expression of both genes in NAC-treated cells. As shown in Figure 1A
, NAC treatment of 308 cells resulted in 3- to 5-fold induction of p21WAF1/CIP1 protein and mRNA. Surprisingly, p21WAF1/CIP1 protein elevation by NAC was also detected in the p53-deficient (10)1 fibroblast cell line (Figure 1A
). These observations indicate that p21WAF1/CIP1 induction by NAC is independent of p53. Moreover, the G1-specific cdk inhibitor p16INK4a was also induced by NAC (Figure 1B
). p16INK4a is a specific inhibitor of cyclin D-dependent kinase, and it is believed to mediate growth suppression by increasing hypophosphorylated pRb, which in turn leads to G1 cell-cycle arrest (22).


View larger version (41K):
[in this window]
[in a new window]
|
Fig. 1. (A) p53 independence of p21WAF1/CIP1 induction by NAC. Western blot analysis of p21WAF1/CIP1 protein induction by 20 mM NAC in 308 cells (upper); northern blot of p21WAF1/CIP1 mRNA in 308 cells after 5 h exposure to NAC (middle); western blot analysis of p21WAF1/CIP1 protein expression after treatment with 20 mM NAC in p53-deficient 10(1) cells (lower). (B) p16INK4a protein induction by 5 mM NAC in 308 cells. Data shown represent similar results from one of two experiments.
|
|
These studies show that NAC upregulates the expression of p21WAF1/CIP1 and p16INK4a. To investigate the contribution of these proteins, as well as p53, to the G1 cell-cycle arrest by NAC, we performed genetic analysis by using gene-null cells. As shown in Figure 2A and B
and Table II
, while p53-null and p21WAF1/CIP1-null cells retained the full G1 arrest, ~40% of the arrest was lost in p16INK4a knockout cells. Therefore, we conclude that the G1 arrest by NAC is partially governed by p16INK4a, but not by p21WAF1/CIP1 or p53.


View larger version (30K):
[in this window]
[in a new window]
|
Fig. 2. p53 independence of G1 arrest by NAC. (A) Profiles of cell-cycle distribution in p53-deficient (10)1 cells treated with NAC for 24 h. (B) Profiles of cell-cycle distribution in mouse keratinocyte cell line NHK-4 (p53/) treated with 5 mM NAC for 24 h. Data shown represent similar results from one of two or three experiments.
|
|
We next investigated whether NAC-induced G1 phase delay could be reversed by removing NAC from the medium. 308 cells were treated with 20 mM NAC for 24 h, and the medium was replaced with medium not containing NAC. As seen in Figure 3
, 24 h after the medium replacement, cells resumed their regular cell-cycle distribution. Thus, the NAC effect on G1 arrest is reversible.

View larger version (15K):
[in this window]
[in a new window]
|
Fig. 3. Reversibility of G1 arrest by NAC. 308 cells were incubated with medium containing 20 mM NAC for 24 h, and replaced with regular medium containing no NAC. Twenty-four hours later, cells were subjected to flow cytometry analysis. Data shown represent similar results from one of two experiments.
|
|
A major intracellular pathway of NAC metabolism is deacetylation to the thiol cysteine, and subsequent incorporation into GSH (13). GSH is the major cellular antioxidant. To test the possibility that NAC acts by increasing the intracellular GSH level, we pre-treated and co-incubated cells with L-buthionine sulfoximine (BSO), a potent inhibitor of GSH synthesis. Significantly, G1 arrest by NAC was not blocked by 20 µM BSO (Figure 4
). This dose of BSO was found to completely block the increase in cellular GSH level after NAC (data not shown). BSO itself has little effect on cell-cycle distribution (Figure 4
). These results imply that NAC prolongs the G1 transition via a GSH-independent mechanism.

View larger version (14K):
[in this window]
[in a new window]
|
Fig. 4 Independence of GSH in G1 arrest by NAC. Cell cycle distributions were analyzed in 308 cells treated with BSO and/or NAC. Cells treated with both compounds were preincubated with medium containing 20 µM BSO for 1 h and then treated with medium containing 20 µM BSO and 5 mM NAC for 24 h. Data shown represent similar results from one of two experiments.
|
|
Since NAC is an antioxidant, we also tested a chain-breaking antioxidant, Trolox, which is a water-soluble analog of Vitamin E. At a dose of 1 mM (the maximal solubility in medium), Trolox did not alter the cell cycle-distribution or cause any G1 arrest (Figure 5
). This result indicates that the G1-phase accumulation is not a general effect of antioxidants and suggests, but does not prove, that the cell-cycle arrest by NAC is mediated by its redox effect rather than free radical scavenging.

View larger version (13K):
[in this window]
[in a new window]
|
Fig. 5. No effect of non-sulfur containing antioxidant Trolox on cell cycle progression. 308 cells were incubated with 1 mM Trolox for 24 h, and then subjected to flow cytometry analysis. Data shown represent similar results from one of two experiments.
|
|
All of the above studies were performed in mouse cells. In order to assess the possibility that G1-phase prolongation may contribute to NAC's chemoprevention of human cancers, we next investigated whether NAC could delay G1 transition in human cells. Human primary foreskin fibroblasts were treated with 1 mM NAC for 24 h, and subjected to flow cytometry analysis. As shown in Figure 6
, G1 arrest indeed occurred in normal human cells. This observation indicates that G1 arrest by NAC is not species specific.

View larger version (15K):
[in this window]
[in a new window]
|
Fig. 6. G1 arrest by NAC in human primary foreskin fibroblasts. Fibroblasts were incubated with 1 mM NAC for 24 h, and then subjected to flow cytometry analysis. Data shown represent similar results from one of two experiments.
|
|
 |
Discussion
|
---|
In this report, we show that NAC treatment upregulates p21WAF1/CIP1 and p16INK4a expression, and prolongs the G1-phase transition in part via p16INK4a. G1 arrest by NAC is independent of p53 and p21WAF1/CIP1. As a chemopreventive agent, NAC has often been reported to exert its chemopreventive activities by blocking the formation of DNAcarcinogen adducts, either directly or indirectly (5). Our current findings unveil a potential mechanism by which NAC may accomplish its chemoprevention function in normal cells by inducing p16INK4a-mediated G1 cell-cycle arrest and p21WAF1/CIP1 gene expression.
As an antioxidant, NAC was demonstrated to inhibit DNA synthesis by ras-transformed fibroblasts, via scavenging reactive oxygen species elicited by activated ras (23). However, the role of reactive oxygen species in normal cells is less clear, since these were not measured (23). If normal mitogenic signaling is mediated by reactive oxygen species, our data suggest that reactive oxygen species act partially by inhibiting p16INK4a and thus relieving G1 prolongation.
In view of our previous report on NAC-induced apoptosis (14), and results presented in this study, it is clear that the fate of 308 papilloma cells after NAC treatment can be modulated by the dose applied. At a dose of 5 mM or lower, G1 arrest occurs with very little apoptosis; at 20 mM, both G1 arrest and apoptosis are induced. In contrast to G1 arrest, however, the apoptosis by NAC is not detected in p53-deficient cells (14). These findings suggest that G1 arrest and apoptosis induced by NAC occur through different pathways, with the G1 arrest acting through a p53-independent mechanism.
In their lifetimes, humans are constantly exposed to low doses of a variety of environmental carcinogens. Since it is impossible to design different chemopreventive agents to inhibit innumerable carcinogens with distinctive mechanisms, it is reasonable to make the hypothesis that one effective strategy for chemoprevention would be to apply extrinsic regulators that facilitate natural cellular defenses against general genetic insults. Activation of tumor suppressor genes, such as p16INK4a and p53 (22,24), fits well with this strategy. Cell proliferation is a risk factor for response to carcinogenic stimuli, such as DNA damage. By slowing down cell proliferation via p16INK4a induction, NAC may facilitate cellular machinery for repairing genomic damage by carcinogens before cell division.
While tumor suppressor genes such as p16INK4a and p53 would function well in normal cells, they can be inactivated through gene alterations during tumorigenesis. Their lost functions may be offset by other genes with anti-tumorigenesis activities, such as p21WAF1/CIP1. Unlike p16INK4a and p53, there are few p21WAF1/CIP1 gene mutations detected in human malignancy (25). p21WAF1/CIP1 has been suggested to play a role in DNA repair and slowdown of DNA replication in S phase (2628), in addition to cell cycle control (19). Therefore, p53-independent p21WAF1/CIP1 activation by NAC could function in both normal and precancer cells.
 |
Acknowledgments
|
---|
We are very grateful to Drs P.Leder, M.Serrano, W.Weinberg and A.Levine for providing transgenic mice and cell lines, respectively. This work was supported by NIH grant CA55737 (D.E.B.) and the Leslie H.Warner Postdoctoral Fellowship in Cancer Research (to M.L.).
 |
Notes
|
---|
3 To whom correspondence should be addressed Email: douglas.brash{at}yale.edu 
 |
References
|
---|
-
Flora,S.D., Astengo,M., Serra,D. and Bennicelli,C. (1986) Inhibition of urethan-induced lung tumors in mice by dietary N-acetylcysteine. Cancer Lett., 32, 235241.[ISI][Medline]
-
Rostein,J.B. and Slaga,T.J. (1988) Anticarcinogenesis mechanism, as evaluated in the multistage mouse skin model. Mutat. Res., 202, 421427.[ISI][Medline]
-
Steele,V.E., Kelloff,G.J., Wilkinson,B.P. and Arnold,J.T. (1990) Inhibition of transformation in cultured rat tracheal epithelial cells by potential chemopreventive agents. Cancer Res., 50, 20682074.[Abstract]
-
Pereira,M.A. and Khoury,M.D. (1991) Prevention by chemopreventive agents of azoxymethane-induced foci of aberrant crypts in rat colon. Cancer Lett., 61, 2733.[ISI][Medline]
-
Flora,S.D., Izzotti,A., D'Agostino,F., Balansky,R. and Cesarone,C.F. (1992) Chemopreventive properties of N-acetylcysteine and other thiols. In Wattenberg,L., Lipkin,M., Boone,C.W. and Kelloff,G.J. (eds) Cancer Chemoprevention. CRC Press, Boca Raton, FL, pp. 183194.
-
Izzotti,A., D'Agostini,F., Bagnasco,M., Scatolini,L., Rovida,A., Balansky,R.M., Cesarone,C.F. and De Flora,S. (1994) Chemoprevention of carcinogenDNA adducts and chronic degenerative diseases. Cancer Res., 54, 1994s1998s.[Abstract]
-
Aruoma,O.I., Halliwell,B., Hoey,B.M. and Butler,J. (1989) The antioxidant action of N-acetylcysteine: its reaction with hydrogen peroxide, hydroxyl radical, superoxide and hypochlorous acid. Free Rad. Biol. Med., 6, 593597.[ISI][Medline]
-
Burgunder,J.M., Varriale,A. and Lauterburg,B.H. (1989) Effect of N-acetylcysteine on plasma cysteine and glutathione following paracetamol administration. Eur. J. Clin. Pharmacol., 36, 127131.[ISI][Medline]
-
Smilkstein,M.J., Knapp,G.L., Kulig,K.W. and Rumack,B.J. (1988) Efficacy of oral N-acetylcysteine in the treatment of acetaminophen overdose. N. Engl. J. Med., 319, 15571562.[Abstract]
-
Solen,G. (1993) Radioprotective effect of N-acetylcysteine in vitro using the induction of DNA breaks as end-point. Int. J. Radiat. Biol., 64, 359366.[ISI][Medline]
-
Yunis,A.A., Lim,L.O. and Arimura,G.K. (1986) DNA damage induced by chloramphenicol and nitroso-chloramphenicol: protection by N-acetylcysteine. Respiration, 50 (suppl.), 5055.[ISI][Medline]
-
Chan,J.Y.H., Stout,D.L. and Becker,F.F. (1986) Protective role of thiols in carcinogen-induced DNA damage in rat liver. Carcinogenesis, 7, 16211624.[Abstract]
-
Holdiness,M.R. (1991) Clinical pharmacokinetics of N-acetylcysteine. Clin. Pharmacokinet., 20, 123134.[ISI][Medline]
-
Liu,M., Pelling,J.C., Ju,J., Chu,E. and Brash,D.E. (1998) Antioxidant action via p53-mediated apoptosis. Cancer Res., 58, 17231729.[Abstract]
-
Strickland,J.E., Greenhalgh,D.A., Koceva-Chyla,A., Hennings,H., Restrepo,C., Balaschak,M. and Yuspa,S.H. (1988) Development of murine epidermal cell lines which contain an activated rasHa oncogene and form papillomas in skin grafts on athymic nude mouse hosts. Cancer Res., 48, 165169.[Abstract]
-
Azzoli,C.G., Sagar,M., Wu,A., Lowry,D., Hennings,H., Morgan,D.L. and Weinberg,W.C. (1998) Cooperation of p53 loss of function and v-Ha-ras in transformation of mouse keratinocyte cell lines. Mol. Carcinog., 21, 5061.[ISI][Medline]
-
Harney,D.M. and Levine,A.J. (1991) p53 alteration is a common event in the spontaneous immortalization of primary BALB/c murine embryo fibroblasts. Genes Dev., 5, 23752385.[Abstract]
-
Dlugosz,A.A., Glick,A.B., Tennenbaum,T., Weinberg,W.C. and Yuspa,S.H. (1995) Isolation and utilization of epidermal keratinocytes for oncogene research. Methods Enzymol., 254, 321.[ISI][Medline]
-
Deng,C., Zhang,P., Harper,J.W., Elledge,S.J. and Leder,P. (1995) Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell, 82, 675684.[ISI][Medline]
-
Serrano,M., Lee,H., Chin,L., Cordon-Cardo,C., Beach,D. and DePinho,R.A. (1996) Role of the INK4a locus in tumor suppression and cell mortality. Cell, 85, 2737.[ISI][Medline]
-
Liu,M. and Pelling,J.C. (1995) UV-B/A irradiation of mouse keratinocytes results in p53-mediated WAF1/CIP1 expression. Oncogene, 10, 19551960.[ISI][Medline]
-
Serrano,M. (1997) The tumor suppressor protein p16INK4a. Exp. Cell Res., 237, 713.[ISI][Medline]
-
Irani,K., Xia,Y., Zweier,J.L., Sollott,S.J., Der,C.J., Fearon,E.R., Sundaresan,M., Finkel,T. and Goldschmidt-Clermont,P.J. (1997) Mitogenic signaling mediated by oxidants in ras-transformed fibroblasts. Science, 275, 16491652.[Abstract/Free Full Text]
-
Ko,L.J. and Prives,C. (1996) p53: puzzle and paradigm. Genes Dev., 10, 10541072.[ISI][Medline]
-
Shiohara,M., El-Deiry,W.S., Wada,M., Nakamaki,T., Takeuchi,S., Yang,R., Chen,D.L., Vogelstein,B. and Koeffler,H.P. (1994) Absence of WAF1 mutations in a variety of human malignancies. Blood, 84, 37813784.[Abstract/Free Full Text]
-
Sheikh,M.S., Chen,Y.Q., Smith,M.L. and Fornace,A.J. (1997) Role of p21Waf1/Cip1/Sdi1 in cell death and DNA repair as studied using a tetracycline-inducible system in p53-deficient cells. Oncogene, 14, 18751882.[ISI][Medline]
-
McDonald,E.R., Wu,G.S., Waldman,T. and El-Deiry,W.S. (1996) Repair defect in p21 WAF1/CIP1/ human cancer cells. Cancer Res., 56, 22502255.[Abstract]
-
Waga,S., Hannon,G.J., Beach,D. and Stillman,B. (1994) The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature, 369, 574578.[ISI][Medline]
Received June 5, 1998;
revised May 5, 1999;
accepted May 14, 1999.