the molecular basis for cell cycle delays following ... advance/journals/radion/ecco37.pdfthe...

13
ELSEVIER SCIENCE IRELAND Radiotherapy and Oncology 31 (1994) I - 13 R ADIOTHERAPY aoNC~~OG~ The molecular basis for cell cycle delays following ionizing radiation: a review Amit Maity”, W. Gillies McKennaa, Ruth J. Muschel*b ‘Department of Radiation Oncology. hDeparrment of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104. USA (Received 15 September 1993: revision received 8 December 1993: accepted 20 December 1993) Abstract Exposure of a wide variety of cells to ionizing (X- or y-) irradiation results in a division delay which may have several components including a G, block, a G2 arrest or an S phase delay. The G, arrest is absent in many cell lines, and the S phase delay is typically seen following relatively high doses (> 5 Gy). In contrast, the G2 arrest is seen in virtually all eukaryotic cells and occurs following high and low doses, even under 1 Gy. The mechanism underlying the G2 arrest may involve suppression of cyclin Bl mRNA and/or protein in some cell lines and tyrosine phosphorylation of ~34”~“’ in others. Similar mechanisms are likely to be operative in the G2 arrest induced by various chemotherapeutic agents including nitrogen mustard and etoposide. The upstream signal transduction pathways involved in the G2 arrest following ionizing radiation remain obscure in mammalian cells; however, in the budding yeast the rud9 gene and in the fission yeast the chkl/rad27 gene are involved. There is evidence indicating that shortening of the G2 arrest results in decreased survival which has led to the hypothesis that during this block, cells repair damaged DNA following exposure to genotoxic agents. In cell lines examined to date, wildtype ~53 is required for the G, arrest following ioniz- ing radiation. The gudd45 gene may also have a role in this arrest. Elimination of the G, arrest leads to no change in survival following radiation in some cell lines and increased radioresistance in others. It has been suggested that this induction of radioresistance in certain cell lines is due to loss of the ability to undergo apoptosis. Relatively little is known about the mechanism underlying the S phase delay. This delay is due to a depression in the rate of DNA synthesis and has both a slow and a fast compo- nent. In some cells the S phase delay can be abolished by staurosporine, suggestinginvolvement of a protein kinase. Understanding the molecular mechanisms behind these delays may lead to improvement in the efficacy of radiotherapy and/or chemotherapy if they can be exploited to decrease repair or increase apoptosis following exposure to these agents. Key words: Radiation; Cell cycle delay; Cyclin ~53; cdc2 1. Background Irradiation of a wide variety of cells, including amoeba [l], sea urchin eggs [2], yeast [3], and mammali- an cells, causes a division delay. In mammalian cells, this delay may have several components including a G1 arrest, an S phase delay and a G2 arrest. Although this * Corresponding author, Room 520, Clinical Research Building, University of Pennsylvania, 422 Curie Blvd., Philadelphia, PA 19104, USA. has heen known for many years, it is only recently that progress in our understanding of cell cycle regulation through checkpoints [4] and feedback controls [5,6], has offered new insight into the underlying biochemical mechanisms. This review will summarize our current knowledge regarding the S phase delay following radia- tion, the role of ~53 in the G1 arrest and the role of cyclins and ~34’~‘~ in the G2 arrest. Much of the earlier work on radiation effects on mammalian cells was done in HeLa cells. By calculating the percentage of mitotic figures (mitotic index) at 0167-8140/94/$07.00 0 1994 Elsevier Science Ireland Ltd. All rights reserved. SSDI 0167-8140(93)01345-P

Upload: others

Post on 25-Feb-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

ELSEVIER SCIENCE IRELAND Radiotherapy and Oncology 31 (1994) I - 13

R ADIOTHERAPY aoNC~~OG~

The molecular basis for cell cycle delays following ionizing radiation: a review

Amit Maity”, W. Gillies McKennaa, Ruth J. Muschel*b ‘Department of Radiation Oncology. hDeparrment of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine,

Philadelphia, PA 19104. USA

(Received 15 September 1993: revision received 8 December 1993: accepted 20 December 1993)

Abstract

Exposure of a wide variety of cells to ionizing (X- or y-) irradiation results in a division delay which may have several components including a G, block, a G2 arrest or an S phase delay. The G, arrest is absent in many cell lines, and the S phase delay is typically seen following relatively high doses (> 5 Gy). In contrast, the G2 arrest is seen in virtually all eukaryotic cells and occurs following high and low doses, even under 1 Gy. The mechanism underlying the G2 arrest may involve suppression of cyclin Bl mRNA and/or protein in some cell lines and tyrosine phosphorylation of ~34”~“’ in others. Similar mechanisms are likely to be operative in the G2 arrest induced by various chemotherapeutic agents including nitrogen mustard and etoposide. The upstream signal transduction pathways involved in the G2 arrest following ionizing radiation remain obscure in mammalian cells; however, in the budding yeast the rud9 gene and in the fission yeast the chkl/rad27 gene are involved. There is evidence indicating that shortening of the G2 arrest results in decreased survival which has led to the hypothesis that during this block, cells repair damaged DNA following exposure to genotoxic agents. In cell lines examined to date, wildtype ~53 is required for the G, arrest following ioniz- ing radiation. The gudd45 gene may also have a role in this arrest. Elimination of the G, arrest leads to no change in survival following radiation in some cell lines and increased radioresistance in others. It has been suggested that this induction of radioresistance in certain cell lines is due to loss of the ability to undergo apoptosis. Relatively little is known about the mechanism underlying the S phase delay. This delay is due to a depression in the rate of DNA synthesis and has both a slow and a fast compo- nent. In some cells the S phase delay can be abolished by staurosporine, suggesting involvement of a protein kinase. Understanding the molecular mechanisms behind these delays may lead to improvement in the efficacy of radiotherapy and/or chemotherapy if they can be exploited to decrease repair or increase apoptosis following exposure to these agents.

Key words: Radiation; Cell cycle delay; Cyclin ~53; cdc2

1. Background

Irradiation of a wide variety of cells, including amoeba [l], sea urchin eggs [2], yeast [3], and mammali- an cells, causes a division delay. In mammalian cells, this delay may have several components including a G1 arrest, an S phase delay and a G2 arrest. Although this

* Corresponding author, Room 520, Clinical Research Building, University of Pennsylvania, 422 Curie Blvd., Philadelphia, PA 19104, USA.

has heen known for many years, it is only recently that progress in our understanding of cell cycle regulation through checkpoints [4] and feedback controls [5,6], has offered new insight into the underlying biochemical mechanisms. This review will summarize our current knowledge regarding the S phase delay following radia- tion, the role of ~53 in the G1 arrest and the role of cyclins and ~34’~‘~ in the G2 arrest.

Much of the earlier work on radiation effects on mammalian cells was done in HeLa cells. By calculating the percentage of mitotic figures (mitotic index) at

0167-8140/94/$07.00 0 1994 Elsevier Science Ireland Ltd. All rights reserved. SSDI 0167-8140(93)01345-P

Page 2: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Maity et al. / Radiother. Oncol. 31 (1994) 1-13

various times, Painter and Robertson [7] showed that in HeLa S3 cells there was a division delay within 2 h following exposure to 500 R which lasted for at least 8 h and disappeared by 14.5 h. By pulse labelling with [3H]thymidine and determining the percentage of la- belled cells, they demonstrated an accumulation of cells in S phase between 4 and 8 h following this dose. Painter [8] subsequently showed that this accumulation resulted from a direct depression in the rate of DNA synthesis while the rate of entry from Gt to S remained un- changed.

Yamada and Puck [9] irradiated HeLa S3 cells with lower doses of radiation (0.34-l .35 Gy). They con- tinuously labelled cells with [3H]thymidine for various lengths of time immediately following irradiation. By plotting the percentage of labelled nuclei versus time, they showed that the duration of S phase was not significantly affected by these doses of radiation, nor was there a block from G, to S phase. In contrast, they saw a drop in the frequency of metaphase within 1 h of irradiation followed by a rise, an overshoot and then return to baseline. The amount of time for the relative metaphase frequency to return to baseline ranged from 2 h for 0.34 Gy to 11 h for 1.35 Gy. Since the frequency started to drop within 1 h after irradiation, they inferred that a block must be occurring within 1 h prior to metaphase, most likely late in G2. They directly demonstrated a G2 block by continuously labelling cells with [3H]thymidine and plotting the percentage of labelled metaphases as a function of time. Using this approach, they found that the duration of G2 delay increased proportionately with dose.

After irradiating HeLa S3 cells synchronized by mitotic shakeoff with 3 Gy during different phases of the cell cycle, Terasima and Tolmach [lo] found that the total division delay was minimal after irradiation in G,, maximal for cells in G2 and intermediate for cells ir- radiated in S. Cells irradiated in G, experienced a slightly prolonged S phase and a minimally prolonged G2 phase. Irradiation in S or G2 resulted in a significant Gz delay. Cells irradiated in S phase also had a pro- longed S phase and a decreased rate of DNA synthesis. No significant G, delay was seen for HeLa cells ir- radiated in any phase.

Yu and Sinclair [ 1 l] found that synchronized Chinese hamster V79 cells showed a dose dependent mitotic delay following 2.35-7.1 Gy which was greatest for cells irradiated in late S phase, least for cells in Gi and inter- mediate for cells in G2. Likewise, Leeper et al. [12] showed that CHO (Chinese hamster ovary) cells syn- chronized by mitotic shakeoff also exhibited a signifi- cant division delay following 1.5-6 Gy which was dose- and cell-cycle-dependent. The later in the cell cycle irra- diation occurred, the greater was the delay (minimal for G, cells; maximal for late S/G2 cells). As with HeLa cells, most of the delay was due to a block in Gz, but

less pronounced delays in progression through Gi or S were detected [ 131.

Similar effects have been observed in mouse L-cells. Whitmore et al. [14] showed that there was a significant division delay after 20 Gy (19 h) or 50 Gy (25 h) which was primarily due to a reversible block in GZ. By using various radiolabelling techniques, Mak and Till [ 151 were able to detect a decrease in the progression of cells from G, to S and a decrease in the rate of DNA syn- thesis with doses as low as 2.2 Gy and 5.8 Gy, respec- tively.

Work has also been done on cells taken directly from human tissue. Lajtha and coworkers [ 16,171 showed that in human bone marrow cells, doses between 5 and 50 Gy resulted in a decrease in the rate of DNA synthesis as well as a block in movement of cells from G, to S whereas doses of 2 and 3 Gy caused only the latter. Little [18] performed experiments on diploid human amnion cells taken from placenta which had an ex- tremely slow turnover. By continuously labelling with [ 14C]thymidine for several days and determining the la- belling index as a function of time, he showed that even doses as low as 0.1 Gy decreased the flow of cells from Gi to S. This decrease was dose-dependent from 0.1 to 3 Gy with an almost complete Gi arrest at 3 and 10 Gy.

It has been known for many years that caffeine can partially abolish the division delay induced by radiation. Treatment with l-2 mM caffeine prior to irradiation reduces both the inhibition of DNA synthesis (S phase delay) and the G2 delay in both CHO [19] and HeLa cells [20]. Boynton et al. [21] showed that S-180 ascites tumor cells grown intraperitoneally in mice underwent a G2 block following whole body irradiation which could be reduced by pretreating the mice with caffeine.

2. G2 arrest in mammalian cells

Many years ago our laboratory became interested in the molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts transfected with various oncogenes. Several lines were created [22] by transfecting with H-rus and myc (2.8, 2.10 and 3.7) or with myc alone (Myc-Ret and MR4). All three lines transfected with H-rus and myc were significantly more radioresistant (Do, 1.68-2.17) than the two transfected with myc alone (De, 1.06- 1.08) which had a sensitivity comparable to the parental cells. The difference in radiosensitivity between 3.7 and MR4 was not due to differences in the induction of double strand breaks or their rejoining [23]. However, the radioresistant lines displayed a much greater Gz delay following irradiation than the sensitive lines.

Su and Little [24] have shown a similar correlation in human diploid cells transfected with SV40 large T anti- gen. The SV40 immortalized lines were more radioresis- tant than the parental lines (Do, 1.65-2.33 vs.

Page 3: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Maity et al. /Radiother. Oncol. 31 (1994) 1-13 3

1.20-1.35) and showed a greater G2 delay following doses ranging from 2 to 8 Gy.

Since making the correlation between radiosensitivity and G2 delay in this rat embryo model system, our lab- oratory has been trying to determine the mechanisms underlying the radiation-induced G2 arrest. In order to describe these studies, it is necessary to first summarize our current knowledge regarding the biochemistry of the G,/M transition (for review see Ref. 25).

2.1. Cyclins, cdc2 kinase and the G/M transition

Entry of cells into mitosis is regulated by mitosis pro- motion factor (MPF), a complex of two proteins, p34’d’2 and cyclin B. In its activated form, MPF phosphorylates key proteins such as histones and nu- clear lamins, which is thought to lead to the events characterizing mitosis such as chromosome condensa- tion and dissolution of the nuclear envelope. In vitro, MPF activity can be assayed by measuring phosphoryla- tion of histone Hl . In order for cells to complete mitosis and enter Gi, cyclin B must be degraded [26]. Beach et al. [27] originally isolated ~34’~~’ from a temperature sensitive mutant of the fission yeast Schizosaccharo- myces pombe which at the restrictive temperature arrested in either G, or late G2. They also isolated the homologous gene in the budding yeast Saccharomyces cerevisiae, cdc28, previously described by Hartwell et al. [28], as well as the human homolog [29]. The cdc2 gene was found to encode a 34-kDa phosphoprotein with kinase activity, often referred to as ~34’~” or simply p34. p34cdc2 has subsequently been shown to be essen- tial for the G2/M transition in mammalian cells, but its role in Gi is less defined. Riabowol et al. [30] showed that inhibition of ~34’~‘~ in rat fibroblasts by microin- jection of antibodies directed against it prevented entry into mitosis.

The cyclins were discovered by Hunt et al. in marine invertebrates as proteins whose levels oscillated with the cell cycle [31]. Two proteins were identified with slightly different molecular weights and temporal patterns of ex- pression; the one whose level rose earlier and fell earlier was called cyclin A and the other cyclin B. These origi- nal cyclins were mitotic cyclins involved in the Gz/M transition; however, subsequently, Gi cyclins have been discovered (for review see Ref. 32).

Cyclins have been cloned from diverse species including Schiz. pombe [33], humans [34], Drosophila [35], alfalfa [36] and rodents [37]. Sequence comparison shows that there is a relatively conserved region called the ‘cyclin box’ located in the middle third of the pro- tein. Motifs within this can be used to separate the A- from B-type cyclins. All eukaryotes examined to date have been found to contain B-type cyclins; however, A- type cyclins have not been found in yeast. There are at least two types of B cyclins, Bl and B2, in Xenopus [38]

and four types in Sac. cerevisiae [39], but the distinction between these is unknown.

The function of cyclin A is less well defined than that of cyclin Bl. Cyclin A appears to have an important role in DNA synthesis as well as in the G2/M transition. In rat embryo Iibroblasts [40] and HeLa cells, microinjec- tion of anti-cyclin A antibody in G, inhibited DNA synthesis. Microinjection of this antibody into HeLa cells in G2 halted progression into mitosis [41]. Consis- tent with these findings, immunofluorescence in primary human fibroblasts shows that cyclin A has a predominantly nuclear localization from S phase on- ward whereas cyclin Bl accumulates in the cytoplasm of interphase cells and only enters the nucleus at the begin- ning of mitosis [42]. Also consistent with its involvement in DNA synthesis, in synchronized HeLa cells, tran- scription of the cyclin A gene starts about 2 h before cyclin Bl [43]. Whereas in HeLa cells, the level of cyclin Bl protein rises in S phase, peaks in early M phase and falls abruptly at the metaphase/anaphase transition [34], the level of cyclin A starts to rise earlier, peaks earlier and falls abruptly in metaphase [43]. Likewise, the his- tone Hl kinase activity of cyclin A occurs earlier than that of cyclin B 1. The exact function of cyclin A in DNA synthesis is unknown; however, it appears to be carried out in complex with ~33’~~, a protein related to ~34’~” which can associate with cyclin A but not cyclin B 1 [44]. There is evidence that the cyclin Alp33caJ complex binds to and phosphorylates ~107, an Rb-like protein [45]. These three proteins have been found in a quater- nary complex with the transcription factor E2F in S phase [46]. Since E2F binding sites are found within promoters of many genes involved in DNA synthesis, such as thymidine kinase and dihydrofolate reductase, this suggests how cyclin A may be involved in DNA syn- thesis.

2.2. Regulation of cdc2 kinase by phosphorylation

The association of ~34’~” with cyclin B is necessary but not sufficient for generation of kinase activity. There are several residues within ~34’~” whose phosphoryla- tion modifies its activity. In Schiz. pombe, it has been shown that Tyrl5 of ~34’~~~ is phosphorylated in late Gz but must be dephosphorylated in order for cells to proceed into mitosis [47]. Thr167 is also phosphory- lated, but this activates MPF [48].

In mammalian cells, the phosphorylation of ~34’~‘~ is also important in regulating its activity. Norbury et al. [49] and Krek and Nigg [50] showed that for both mouse and chicken ~34 cdc2 there exists a Thr14 as well as a Tyrl5 whose phosphorylation negatively regulates kinase activity. Both residues become dephosphorylated as cells enter mitosis. In mammalian ~34’~‘~, Thr167 is

Page 4: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

4 A. Maity et al. / Radiother. Oncol. 31 (1994) 1-13

the homolog of the Schiz. pombe Thr161 and regulates kinase activity via phosphorylation [51-531.

The exact timing of the phosphorylation of these resi- dues on ~34’~‘~ relative to the binding of cyclin Bl to p34cdc2 is at present uncertain. It appears that phos- phorylation of Tyr15 and Thr14 requires cyclin binding [54]. Results in Schiz. pombe [48] and human ~34’~‘~ [51] suggest that cyclin binding occurs after Thr161/167 is phosphorylated; however, other data suggest that phosphorylation occurs after cyclin binding [53,55]. One model [55] for mammalian cells is that cyclin B binds to unphosphorylated ~34’~” in S phase and induces the phosphorylation of all three residues. The p34cdc2/cyclin Bl complex is kept inactive until Thrl5 and Thr14 are dephosphorylated at the G2/M transition at which time the complex is activated. At the metaphase/anaphase transition, cyclin Bl is degraded and Thrl61 on ~34’~” is dephosphorylated.

A number of regulators of ~34’~‘~ phosphorylation have been identified including the phosphatase cdc25 and the kinases wee1 and mikl. cdc25 was originally cloned by Russell and Nurse [56] from Schiz. pombe, and several years later the human homolog was cloned [57]. cdc25 encodes a phosphatase which specifically dephosphorylates Tyrl5 in lower eukaryotes and both Tyrl5 and Thr14 in higher eukaryotes [58,59]. The phosphatase activity of cdc25 is itself regulated by phos- phorylation, increasing immediately prior to mitosis [60]. Hoffman et al. [61] showed that in human cells cdc25 is hyperphosphorylated near mitosis at the time of maximal phosphatase activity. They also showed that this form can activate the p34cdc2/cyclin Bl complex. Interestingly, this activated complex can phosphorylate cdc25, raising the possibility of an autocatalytic feed- back loop.

Another mitotic regulator, wee1 was cloned by Russell and Nurse [62] by complementation of a yeast mutant that entered mitosis prematurely. This gene en- codes a 107-kDa dual specificity protein which can phosphorylate both tyrosine and serine residues [63]. The human homolog of wee1 has been cloned [64] and found to encode a 49-kDa protein. Parker and Piwnica- Worms [65] showed that in vitro, human p49’““’ catalysed the phosphorylation of Tyr 15 but not Thr14 of p34cdc2/cyclin Bl. This resulted in an inhibition of his- tone Hl kinase activity which was partially prevented by the addition of cdc25. McGowan and Russell [66] also have shown that wee1 can phosphorylate Tyrl5 but not Thr14 of ~34’~” in the MPF complex. Furthermore, overexpression of human wee1 in HeLa cells by trans- ient transfection led to an arrest in G2 without affecting the increase in cell volume. In Sac. cerevisiae, wee1 activity is negatively regulated by phosphorylation at its C-terminus end by another kinase, niml (new inducer of mitosis)/cdrl (changed division response) [67].

A second gene, mikl (mitosis inhibitory kinase) was cloned by Lundgren et al. [68] by complementation of another Schiz. pombe mutant which also entered mitosis prematurely. These investigators showed that wee1 and mikl have redundant functions. Deletion of both resulted in tyrosine dephosphorylation of ~34’~‘~ and premature lethal entry into mitosis.

Both cdc25 and wee1 are thought to be important in coupling mitosis and cell division to earlier events in the cell cycle. Schiz. pombe mutants which overexpress cdc25 undergo premature cell division, resulting in cells smaller than wildtype. Hydroxyurea, which inhibits DNA synthesis, halts cell cycle progression in wildtype cells; however, in these cdc25 overexpressors, cell divi- sion continues, resulting in anucleate cells or cells con- taining fragments of chromosomal material [69]. In contrast, weel-50 mutants which lack functional wee1 and also divide prematurely are arrested following expo- sure to hydroxyurea. Based on these results, it has been suggested that cdc25 but not wee 1 is important in coupl- ing cell division to the completion of DNA replication. Because overexpression of wee1 results in a dose- dependent delay in mitosis until cells have grown to a larger size than normal, it was thought that perhaps its function might be to coordinate cell division to cell size. More recent work [70] suggests that this model may be too simplistic as there are situations in which the DNA replication checkpoint is still intact in spite of the absence of cdc25 and other instances in which overex- pression of wee1 can restore it.

2.3. Role of cyclins and cdc2 kinase in G2 arrest

Because cyclin Bl is required for the G2/M transi- tion, our laboratory decided to examine the effects of irradiation on its mRNA and protein [71]. HeLa cells were originally chosen for the study because at the time these investigations were initiated, the human cyclin Bl cDNA had been cloned [34]. HeLa cells were syn- chronized at the G&S boundary using a sequential thymidine/aphidocolin block. Shortly after release from the block, the cells were irradiated with 10 Gy, and samples were taken at serial timepoints from replicate plates for flow cytometry and RNA analysis. The ir- radiated cells showed a delayed entry into Gz compared with control unirradiated cells, consistent with the previously described S phase delay 171. However, once in Gz, the irradiated cells stayed for at least 9 h longer than the controls. The interesting observation is that cyclin Bl mRNA levels remained low during this ac- cumulation in GZ. In contrast, in the control cells, cyclin Bl mRNA rose ten-fold in G2 and then fell as cells progressed into Gt. This led to the hypothesis that suppression of cyclin Bl mRNA was partly responsible for the Gz arrest after irradiation in S phase. Another

Page 5: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Muiry el al. / Rudiotlwr. Oncol. 31 (1994) 1-13

experiment was performed in which HeLa cells were synchronized in G,/S, released and then irradiated with 6 Gy in late S/early G,. Again, the irradiated cells showed a G2 delay, taking 7.5 h to enter Gi compared with 2.5 h for the control cells. Northern blotting show- ed that the cyclin Bl mRNA did rise and peak in a pat- tern similar to that seen in control cells. However, Western blotting showed that the level of cyclin Bl pro- tein was suppressed for 4-5 h following radiation. In contrast, in the control cells, cyclin Bl protein peaked within 1 h of time zero and then fell within 3 h, cor- responding to an exit of cells into Gi. Therefore, radia- tion appears to be able to downregulate cyclin Bl expression in HeLa cells at the mRNA or protein level depending on the phase of the cell cycle in which they are irradiated.

In more recent experiments, Muschel et al. [72] have shown that the suppression of cyclin Bl mRNA with ir- radiation of HeLa cells in S phase is dose dependent and can be detected after as little as 2 Gy. Using similar syn- chrony experiments as described above, the effects of ra- diation on cyclin A mRNA and protein were also analysed. In contrast to cyclin Bl, cyclin A mRNA started to increase in irradiated cells at the same time as in control cells. As cells accumulated in G2/M in the ir- radiated cells, the cyclin A mRNA level continued to rise and peaked at a level higher than that seen in con- trol cells. Likewise, the cyclin A protein levels also over- shot the levels seen in control cells. It is unknown whether this overshoot is related to the supression of cyclin B or whether it is part of the mechanism of the Gz delay.

Datta et al. [73] examined the effects of radiation on cyclin Bl expression using a human myeloid leukemia cell line, U-937. They found that a population of cells enriched in G, by elutriation when irradiated with IO Gy showed a decrease in cyclin B mRNA expression with time but a progressive increase in cdc2, cyclin A and cdc25 mRNA expression. A similar decrease in cyclin Bl mRNA expression was seen following irradia- tion of S phase enriched cells with 10 Gy.

Lock and Ross [74] investigated the effects of radia- tion on CHO cells and found that exposure of asyn- chronously dividing cells to 8 Gy resulted in an accumulation of cells in Gz and a rapid inhibition of P34’Jc2 histone Hl kinase activity within I h. Uckun [75] found that irradiation of Ramos Burkitt’s lympho- ma and K562 erythroleukemia cells with l-8 Gy resulted in enhanced tyrosine phosphorylation of P34 cJc2 and an inhibition of its histone Hl kinase activ- ity which correlated temporarity with the Gz delay. Furthermore, pretreatment with herbimycin. a known inhibitor of protein tyrosine kinases, prevented this inhibition of kinase activity as well as the G, delay.

As mentioned previously, caffeine has long been

s

known to abolish both the S phase delay and the G? arrest induced by radiation. Hain et al. [76] showed that in V79 hamster cells, incubation in 4 mM caffeine 4 h after exposure to 7 Gy reversed the accumulation of cells both in S and Gz phase. An explanation of this was suggested by the fact that ~34”~“’ kinase activity in- creased by a factor of four within 30 min of caffeine treatment.

A number of chemotherapeutic agents can also induce a Gz delay. Lock and Ross [74] found that CHO cells exposed to the epipodophyllotoxin etoposide underwent a Gz delay during which ~34”“~ histone Hl kinase activity was inhibited. More recently, Lock [77] has shown that the cells arrested in Gz following exposure to etoposide had a persistent tyrosine phosphorylation of ~34’~” which could explain their loss of histone Hl kinase activity. A similar correlation between G2 delay and persistent tyrosine phosphorylation of ~34”‘“” has been observed in HeLa cells following exposure to the drug camptothecin, a DNA topoisomerase I inhibitor [78], and in CA46, a human lymphoma line, following exposure to nitrogen mustard [79]. The persistent hyper- phosphorylation of ~34”“” in human lymphoma cells after exposure to nitrogen mustard is accompanied by a failure of cdc25 to be converted to its active hyper- phosphorylated form [80]. O’Connor et al. [81] recently showed that the nitrogen mustard-induced G2 delay in CA46 cells correlates with a suppression of cyclin Bllp34”““’ and cyclin Alp34”““’ kinase activity but not cyclin A/p33cdk2 kinase activity. Furthermore, pentox- ifylline, a methylxanthine related to caffeine which has been shown to enhance cytotoxicity of alkylating agents [82], abolished the delay and reverted the cyclin A and Bl kinase activity to that seen in controls.

It has been hypothesized that the Gz arrest following DNA damage may also serve the function of allowing cells to repair damage. Tobe.y [83] speculated that ‘a surveillance mechanism operates throughout G, to eliminate from the proliferative mode cells with altered DNA. Such a surveillance mechanism would examine cells at multiple stages within G, to ensure that either the damaged DNA was repaired normally, in which case a cell would be returned to cycle, or a unrepaired cell would be converted to a non-viable state’. There is no direct evidence for this hypothesis; however, there is some circumstantial evidence. As discussed above, in- vestigators have made correlations between the length of G2 and radiosensitivity [22,24]. Caffeine, which abrogates the G2 delay following radiation, decreases cell survival (841 although this is difficult to interpret since the drug has many effects. Likewise, pentoxifylline has also been shown to sensitize several cell lines in cul- ture to radiation [85].

Arrest in G2 following exposure to radiation and various other DNA damaging agents is seen in diverse

Page 6: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

6 A. .&laity et al. / Radiother. Oncol. 31 (1994) I-13

cell types; however, the actual mechanism may vary. In irradiated HeLa cells, the Gz arrest has been associated with downregulation of cyclin B at either the RNA or protein level. However, in other systems using different cells and/or agents, alterations in ~34’~‘~ phosphoryla- tion have been described. All of these changes have a common endpoint: the inactivation of MPF, which prevents entry into mitosis. The basis for these varia- tions remains unclear. It is possible that the same cell type may use different mechanisms depending on the agent to which it is exposed. For example, in HeLa cells, changes are seen in cyclin B expression following radia- tion but in ~34’~‘~ phosphorylation following exposure to camptothecin. It is noteworthy that some of the cell types on which these studies have been performed are derived from lymphocytes, which, unlike most cells, undergo a mitotic death mediated by apoptosis rather than a postmitotic interphase death. It is not known whether this has any bearing on the mechanism of G2 delay.

It is clear we have just started to outline the mechanisms responsible for the Gz arrest following DNA damage. It needs to be established whether DNA damage is actually repaired during the Gz arrest and whether modulation of the duration of this arrest can alter cell viability. The details regarding detection of this damage and transduction of this signal to ultimately inactivate MPF are also obscure. Since it appears that different pathways can be used to achieve the same end, it remains to be determined how cells choose a certain pathway over another.

3. G2 arrest in yeast

Yeasts, which have proven so useful in understanding the genetics of cell cycle regulation, have also been helpful in gaining insight into the genetic basis for the G, delay following radiation. Much of the pioneering work in this area was done by Weinert and Hartwell [86] who focused their attention on the budding yeast Sac. cerevisiae in which individual cells can be followed easily through the cell cycle by changes in nuclear morphol- ogy. They isolated a particular mutant, rad9, which lacked the ability to arrest in G2 following X- irradiation. A dose of 20 Gy, which killed 68% of raa’!9 mutants, caused G2 arrest in only 18% of the cells. The same dose killed 52% of RAD+ (wildtype) cells but ar- rested 49% of the cells. They then performed experi- ments with yeast cells synchronized in G2 by using the microtubule blocking agent methylbenzamide-2-y-l- carbamate (MBC). They irradiated the cells with 0, 20 or 80 Gy, removed the MBC and replated the yeast on fresh plates. RAD+ cells showed a dose-dependent division delay which was much greater than that seen in rad9 mutants. They then irradiated cells synchronized in G, with MBC and left them in the drug in order to

impose a drug-induced delay. rad9 mutants held in MBC for 4 h after irradiation showed dramatic improvement in cell viability, not seen in RAD+ cells.

Weinert and Hartwell [87] subsequently cloned the ra& gene by complementation and found it to encode a protein with no similarity to any known proteins. rad9 mRNA is constitutively expressed by cells and not induced by irradiation. A ra&null mutant which com- pletely lacked the gene did not display a Gz arrest following irradiation. However, in the absence of irradi- ation, this mutant exhibited normal growth and cell cycling although it had a spontaneous rate of chromo- somal loss and recombination 7-21-fold greater than the wildtype. Based on these results, Weinert and Hart- well have proposed that rad9 performs a surveillance function along the lines of Tobey’s hypothesis [83] checking on DNA integrity and signalling the cell to arrest in G2 in the event that damage is detected.

Weinert [88] has isolated five other genes, radl7, rad24, and mecl-mec3 (mitosis entry checkpoint) which are required for G2 arrest following radiation. mecl and mec2 also appear to be important in sensing unreplicated DNA since in their absence, cells fail to arrest following exposure to hydroxyurea.

Much work has also been done on the fission yeast Schiz. pombe in which the septation index (% of cells with forming septa) can be used as a measure of division delay. Rowley et al. (891 used the weel-50 mutant which at the restrictive temperature lost wee1 function. At the permissive temperature, these mutants behaved like the wildtype following irradiation with a transient decrease in the septation index; however, at the restrictive tem- perature, there was no division delay. Furthermore, this loss of Gz delay was accompanied by poorer survival, supporting the notion that the G2 delay is important for DNA damage repair and viability. In contrast, a mutant which lacked functional mikl, which, as discussed previously, has overlapping function with wee1 [68], did not lose the radiation-induced G2 delay. The in- vestigators also irradiated mutants which overexpressed cdc25. As expected, these cells also displayed a reduced G2 delay compared with the wildtype. Further differ- ences between these mutants were highlighted by expo- sure to hydroxyurea. As shown previously by Enoch and Nurse [69], weel-50 mutants underwent cell cycle arrest following exposure to hydroxyurea; however, the mutants overexpressing cdc25 showed no delay in cell cycle progression, and mikl-null mutants showed a reduced delay. This suggested that wee1 and mikl operate in two different pathways, the former monitor- ing radiation-induced DNA damage and the latter mon- itoring DNA replication. However, wee1 may not be essential for the radiation-induced G2 arrest in all Schiz. pombe since weel-null mutants have been recently described which can still arrest following DNA damage El.

Page 7: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Maity et al. / Radiother. Oncol. 31 (1994) l-13

Other genes have been identified in Schiz. pombe that appear to function as checkpoints following UV irradia- tion, specifically radl, 3,9 and 17 [90]. Yeast containing mutations of any of these four genes fail to arrest after exposure to either UV irradiation or hydroxyurea and display an extreme sensitivity to both. radl and rad3 have also been shown to be necessary for the G2 arrest following y-irradiation [9 1,921. By screening Schiz. pombe for hydroxyurea sensitivity, Enoch et al. [70] identified five other genes, husl-hus5 (hydroxyurea sen- sitive) which also appear to function as checkpoints. Mutants of all five of these also showed extreme sensi- tivity to W irradiation. The relationship between these nine genes is unknown; however, it is hypothesized that they act in a pathway upstream of weel, mikl and cdc25, monitoring both DNA replication and damage.

Recently Walworth et al. (931 isolated a gene, chkl (checkpoint kinase), from Schiz. pombe which encodes a putative serine/threonine kinase and whose overexpres- sion in wildtype yeast caused a mitotic delay. Disruption of this gene causes failure to arrest in G2 following UV irradiation, rendering cells extremely radiosensitive. These chkl-disrupted mutants do arrest after exposure to hydroxyurea, indicating that this gene is not involved in the pathway sensing DNA replication. Carr et al. (un- published data) independently isolated a gene from Schiz. pombe whose deletion results in loss of the G2 checkpoint following ionizing radiation. This gene was named rad27; however, it has since been shown to be identical to chkl. Based on these observations, it has been suggested that in Schiz. pombe, chkllrad27 couples the DNA damage sensing pathway to the cdc2- dependent cell cycle pathway.

The work done in yeast supports the idea that the G2 delay following radiation is an important determinant in cell survival. In both the fission and budding yeast, mutants which are unable to arrest in Gz consistently show a poorer survival following radiation than the wildtype. In some respects, more progress has been made in defining the genetics of the G2 delay in yeast than in mammalian cells. This is to be expected given the relative ease of genetic manipulation in the former com- pared with the latter. It is hoped that many of the yeast genes will turn out to have structurally or functionally homologous counterparts in mammals so that com- plementation or hybridization techniques can be used to isolate the corresponding mammalian genes.

4. Gl arrest

As discussed in section 1 above, a G1 arrest is not seen in all cell lines following radiation. Recent work suggests that its presence depends on the ~53 status of the cell line. Kastan et al. [94] showed that irradiation of ML-l myeloblastic leukemia cells with 0.5-4 Gy caused an accumulation of cells in both Gs,t and G2/M,

1

consistent with two different blocks. The G, arrest predominated at lower doses and the G2 arrest at higher doses. Cells irradiated in S phase were able to progress into Gz/M. Because of the known anti- proliferative effects of ~53 which will be further describ- ed below, these investigators sought to determine whether there was any correlation with changes in its ex- pression. Indeed, the ~53 protein level rose l-2 h after irradiation and required 48-72 h to return to normal. Northern blots showed no change in ~53 mRNA, sug- gesting that the elevation in protein levels was due to a posttranslational increase in stability.

The responses of a number of other hematopoietic 1941 and non-hematopoietic [95] cell lines (human libro- blasts, colorectal carcinoma cells, osteosarcoma cells) were noteworthy. In general, cell lines which, like ML- 1, express wildtype ~53 exhibited a radiation-induced G, arrest but not lines expressing mutant ~53 or lacking ~53 altogether. In contrast, the G2 arrest following irradia- tion remained intact regardless of ~53 status. Further- more, transfection of mutant ~53 into RKO cells which express wildtype ~53 attenuated the Gi arrest. Con- versely, transfection of wildtype ~53 into HL60, a cell line lacking ~53 and not showing radiation-induced G, arrest, resulted in a partial restoration of the arrest.

More recently, Kastan et al. (961 have investigated the relationship between ~53 and radiation-induced arrest using fibroblasts from ~53 ‘knockout’ transgenic mice in which both wildtype pS3 genes have been deleted using the technique of homologous recombination. These mice develop normally except for an increased frequen- cy of certain tumors, specifically lymphomas and sar- comas [97]. Fibroblasts cultured from these mice were used in experiments in early passage while they still had a normal karyotype and morphology. Fibroblasts from normal mice with both intact ~53 genes showed a mark- ed G, arrest after 2 or 4 Gy. In contrast, libroblasts from mice lacking both ~53 genes showed no arrest, and libroblasts from mice with only one intact ~53 gene showed an intermediate response.

The authors then investigated the possible role of gadd45 in the G, arrest. The gadd (growth arrest and DNA damage-inducible) genes are a set of five genes [98] whose transcription in CHO cells is markedly in- creased by exposure to DNA damaging agents such as the alkylating agent methylmethanesulfonate (MMS) or by growth cessation signals such as serum starvation. One of these genes, gadd45, has been found in human cells and is distinct from the other gads genes in that it can also be strongly induced by X-irradiation [99]. Doses as low as 2 Gy cause a 3-4-fold increase in gadd45 mRNA several hours following irradiation. Kastan et al. [96] found an excellent correlation between gaaV45 mRNA induction and ~53 status. Cell lines con- taining both intact ~53 genes showed a 2-lo-fold increase in gadd45 mRNA 4 h after 20 Gy whereas lines

Page 8: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

8 A. Moiry ct ul. / Rudiothur. Onwl. 31 (1994) I-13

with abnormal ~53 function showed no induction dur- ing this time period. Cells from patients with ataxia- telangiectasia which lacked the ~53 response to radia- tion also showed defective induction of g&d45 mRNA. The authors went on to analyse the DNA sequence of gadd45 and found that the third intron of the gadd45 contains a sequence with a high degree of homology to a previously described ~53 binding sequence [ 1001. They showed that an oligomer containing this sequence could bind wildtype but not mutant ~53 and that when this oligomer was placed upstream of a CAT (chloramphen- icol acetyltransferase) reporter gene, transcription could be activated by wildtype ~53. Based on these results, the authors speculated that gadd45 may act downstream of ~5.3 in a signal transduction pathway necessary for the G, arrest, but this remains to be proven.

The relationship between ~53 and the G, arrest has been confirmed by O’Connor et al. [IO11 who found that they could correctly predict ~53 status (wildtype or mu- tant) in 15 of 17 Burkitt’s lymphoma and lym- phoblastoid lines by determining whether they exhibited an intact G, arrest following 6.3 Gy. The two cell lines which had a reduced ability to arrest in Gi despite both p53 genes being intact turned out by Western blotting to have a blunted induction of ~53 protein following ra- diation.

These results suggest that many established cell lines fail to show a Gi arrest following radiation because they lack wildtype ~53. For example, HeLa cells, which were originally derived from a human cervical car- cinoma, have low levels of ~53 as a result of infection with human papillomavirus which expresses the E6 pro- tein which targets ~53 for destruction [ 1021.

Although the suggestion was made that the G, arrest might be important for enhanced DNA repair, Kastan’s group has been unable to demonstrate that the loss of this arrest increases radiosensitivity. Fibroblasts from ‘~53 knockout’ mice with either one or two copies of the gene deleted showed no difference in radiosensitivity compared with those taken from control littermates, and transfection of mutant ~53 into RKO cells did not alter radiosensitivity [ 1031.

In fact, in some settings, mutant ~53 and loss of G, arrest is associated with increased radioresistance. Lee and Bernstein [104] found that bone marrow cells taken from transgenic mice which contained mutant ~53 displayed &gnificant resistance following -r-irradiation compared with cells from wildtype httermates (D,, 2.48-2.80 vs. 1.75 Gy). O’Connor et al. [ 1011 found that in their lymphoma cell lines, loss of wildtype ~53 and Gi arrest was associated with radioresistance although this was not an absolute correlation.

It is difficult to reconcile these seemingly contradic- tory results; however, an observation made in thymo- cytes may help to clarify this dilemma. Wildtype thymocytes are exquisitely sensitive to doses as low as I

Gy, showing a dramatic increase in ~53 level within 1 h of radiation and undergoing an apoptotic cell death [105]. In contrast, thymocytes from homozygous null ~53 mice with both copies of the gene deleted are extremely resistant to radiation-induced apoptosis [105,106] with doses as high as 20 Gy in spite of a normal apoptotic response when exposed to glucocortic- oids. Thymocytes from heterozygous mice with one intact and one deleted copy of thep53 gene display inter- mediate sensitivity.

It has been suggested [103] that in some cell types such as lymphocytes and bone marrow cells, ~53 is im- portant for triggering both apoptosis and a Gi arrest following radiation but that in others such as fibroblasts it is important only in initiating a Gi arrest. In the former case, loss of wildtype ~53 may lead to decreased apoptosis which is manifested as increased radio- resistance. However, in the latter case in which the ~53 signal is not important for apoptosis, loss of wildtype ~53 function may not affect radiosensitivity.

The work cited above has been extremely helpful in elucidating the function of ~53 within the cell. In spite of intense investigation for over a decade, surprisingly little is known in this regard. Originally discovered to complex with simian virus 40 (SV40) large T antigen in cells transformed with this virus [ 107,108], wildtype ~53 has been shown to have a potent anti-proliferative effect on cells using a variety of assays. Introduction of wildtype ~53 into rat embryo fibroblasts [log] and cell lines derived from osteosarcomas [l lo], colorectal car- cinomas [l 1 l] and glioblastomas [112] arrests cells in G, and prevents entry into S phase. Its introduction into Saos-2, a human osteosarcoma cell line lacking endogenous ~53, decreases tumorigenicity as assayed by subcutaneous injection into nude mice and colony for- mation in soft sugar [I 131.

Mutations in ~53 have been found in diverse sporadic (non-familial) tumors [114] and in fact are the most common genetic change found in human neoplasias [115]. The Li-Fraumeni syndrome [116] which results in the autosomal dominant inheritance of certain tumors, most notably sarcomas and breast carcinomas, often involves a germ-line mutation in ~53 [117]. Some of these mutant ~53 proteins can act as cooperating oncogenes, that is when cotransfected into rat embryo tibroblasts with the rus oncogene, they greatly increase the efficiency of transformation.

There is mounting evidence that ~53 can function as a transcriptional factor [118-1201. More recently, investigators have determined that wildtype ~53 can bind to specific DNA sequences in vitro [ 100,121,122]. Templates containing multiple copies of a 33-bp ~53 binding elements upstream of a CAT reporter gene can be transactivated by wildtype ~53 but not mutant ~53, both in vivo [ 1231 and in vitro [ 1241. Furthermore, addi- tion of mutant ~53 can inhibit transactivation by

Page 9: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Muity em al. / Radiorhur. Oncol. 31 (1994) 1-13 9

wildtype ~53. In spite of the ability of ~53 to activate transcription of genes in experimental constructs, only a few genes have been found to contain naturally occurr- ing ~53 binding sequences: the muscle creatine kinase gene [125], the gadd45 gene [96] and the mdm2 gene [126]. In fact, ~53 has been shown to repress transcrip- tion from many more genes including c-fos, c+n, fl- actin, hsp 70, interleukin 6 and ~53 itself [127,128] through an unknown mechanism.

The ~53 protein does not seem to play a critical role in normal cell growth and differentiation since ‘knockout’ mice in which both wildtype genes have been deleted develop normally [97]. However, these mice do develop certain types of tumors prematurely. This may be related to the observation that in some model systems loss of ~53 can lead to genomic instability [ 129,130]. Normal diploid libroblasts [129] undergo cell cycle arrest when exposed to the drug N-(phosphonacetyl)-L- aspartate (PALA) and display an undetectable level of gene amplification (< 109). In contrast, fibroblasts from ~53 ‘knockout’ mice in which both genes are deleted continue to cycle and display a high level of gene amplification upon exposure to PALA. Taking these findings together with the results relating to radiation, one could speculate that ~53 does not play an important role in the normal day-to-day activity of the cell. How- ever, in the’ presence of damage caused by agents such as radiation, ~53 may be important in either signalling cells to undergo apoptosis or repair the damage in order to prevent genomic instability and subsequent tumori- genesis. Thus, ~53 may serve as a ‘guardian of the genome’ [ 13 I] or ‘molecular policeman’ [ 1321. Although an interesting hypothesis, it remains unproven. Also, it does not explain why in those cell lines in which the ~53 signal initiates a G, arrest but not apoptosis following radiation, loss of wildtype ~53 function does not seem to increase radiosensitivity.

5. S phase delay

Irradiation also causes a slowing of progression through S phase, felt to be due to a depression in the rate of DNA synthesis. This depression of DNA synthesis displays a biphasic dose response with a steep (radiosen- sitive) component at lower doses and a shallow (radioresistant) component at higher doses. Using autoradiography in the presence of t3H]thymidine, Watanabe [133] was able to measure the length of elongating DNA fibers in mouse leukemia L5178Y cells. He found that the rate of DNA chain growth was reduced linearly when plotted against radiation dose with a Do of 130 Gy. Therefore, he postulated that the shallow (radioresistant) component of the dose response curve was due to inhibition of DNA chain elongation. Subsequently, others have shown that the steep (radiosensitive) component of the dose response curve is

due to inhibition of replicon initiation. Makino and Okada [134] pulse-labelled L5178Y cells with [‘Hlthymidine and then performed alkaline sucrose gradient sedimentation of the DNA. Using this tech- nique, they conclusively showed that 5 Gy caused a decrease in the rate of DNA synthesis. Furthermore, they found a deficiency in smaller sized DNA’s up to 2 h following radiation compared with non-irradiated controls, consistent with an inhibition of replicon initia- tion rather than chain elongation. The same technique has been used to show that doses from 5 to 8 Gy cause an inhibition of replicon initiation in CHO. HeLa and mouse L cells [135-1371.

Based on estimates of target size, Painter and Young [136] hypothesized that a single radiation hit blocks the initiation of entire clusters of replicons, not just one replicon. Povirk and Painter [ 1381 irradiated HeLa and bovine kidney cells which had been incubated in media containing 5-bromodeoxyuridine to allow for substitu- tion of approximately 20% of the thymine residues with BrdU. They then irradiated the cells with 313 nm light which produces strand breaks with high efficiency in BrdU-substituted DNA but has little effect on un- substituted DNA. Because this led to a suppression of replicon initiation, they inferred that the target responsi- ble was either the DNA itself or something in the imme- diate vicinity.

Wang and Iliakis [ 1391 examined the effect of radia- tion on DNA synthesis in the oncogene transfected rat embryo fibroblast system described previously. Both the radioresistant 3.7 line (myc and ras transfected) and the radiosensitive MR line (myc transfected) showed the ex- pected biphasic curve when the rate of DNA synthesis was plotted versus dose; however, the inhibition of replicon initiation for the 3.7 cells was much greater than the MR cells. Furthermore, in time course experi- ments, the inhibition of the rate of DNA synthesis in the 3.7 cells was much more prolonged than that seen in the MR cells. Similar experiments were carried out on lines transfected with other oncogenes and in general, lines transfected with ras and myc or ras and adenovirus Ela showed increased inhibition of DNA synthesis com- pared with those transfected with myc or Ela alone, leading to the speculation that t-as is involved in the sig- nal transduction pathway controlling inhibition of DNA synthesis.

More recently, Wang et al. [ 1401 used alkaline sucrose gradient sedimentation to show that in 3.7 cells there was a strong suppression of replicon initiation 1 h after radiation which persisted for up to 6 h. In comparison, MR4 cells showed a much weaker suppression of replicon initiation and a gradual recovery 3-6 h later. No significant differences were seen in inhibition of chain elongation in the two lines. When synchronized 3.7 cells were irradiated in early S phase, they were found to have an S phase delay two to three times longer

Page 10: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

IO

than MR cells using doses from 10 to 50 Gy. Irradiation of 3.7 cells with 10 nM staurosporine, a protein kinase inhibitor, for 3 h showed a marked reduction in the inhibition of DNA synthesis to the levels seen in MR cells. By alkaline sucrose gradient sedimentation, staurosporine was shown to reduce the inhibition of replicon initiation induced by radiation.

Much less is known about the mechanisms underlying the S phase delay than the Gi or G2 arrests. It has been pointed out [141] that in Schiz. pombe, the rad check- point genes may also regulate DNA replicon initiation in response to radiation since none of the rad mutants shows an S phase delay even after doses as high as 600 GY.

6.. Conclusion

Much progress has been made since the classic studies of Tolmach, Painter and others which originally describ- ed the effects of radiation on cell cycle progression. The field of altered cell cycle regulation following radiation and other genotoxic agents is now of great interest to a wide range of researchers in fields extending beyond radiobiology. Much of this interest stems from the fact that discoveries made in this area may ultimately have very practical applications, particularly in the treatment of cancers. If, in fact, following radiation or chemother- apy, cells survive by repairing DNA damage during a G2 arrest, it may be possible to increase cell kill by administering agents which force them through this block prematurely. If mutation of p53 is important in decreasing apoptosis in some tumors following radia- tion, this offers another potential means of manipulating radiosensitivity.

Investigations into cell cycle regulation may also help promote advances in cancer prevention by increasing our understanding of how radiation and various car- cinogens lead to malignancy. It is possible that loss of checkpoint control in non-malignant cells plays a role in the transformation process by allowing them to divide in the face of damaged DNA and accumulate chromo- somal abnormalities. Understanding the mechanisms behind this could be of great importance in preventing cancers in the general population, and specifically sec- ond malignant neoplasms in patients treated with radia- tion and/or chemotherapy.

7. Not added in proof

Since this manuscript was submitted several recent publications have added to our understanding of how p53 leads to a Gl arrest. El-Deiry et al. (Cell 75:817-825, 1993) found that p53 results in the transcriptional activation of the WAF-l/ZIP-1 gene. Harpor et al. (Cell 75:805-816, 1993) have showed that WAF-l/CIP-1 forms complexes with the Gl cyclin-edk

A. Maity et al. /Radiother. Oncol. 31 (1994) I-13

complexes which markedly inhibits their kinase ac- tivities.

8. References

I

5

6

I

8

9

IO

11

I2

13

I4

IS

16

I7

I8

19

20

21

22

Daniels, E. Cell division in the giant amoeba, Pelomyxa carolinesnsis following X-irradiation. J. Exp. Zoo]. 127: 427-461, 1952. Miwa, M., Yamashita, H. and Mari, K. The action of ionizing rays on sea urchin. Gann 33: ‘117, 1939. Bums, V.W. X-ray induced division delay of individual yeast cells. Radiat. Res. 4: 394-412, 1956. Hartwell, L.H. and Weinert, T.A. Checkpoints: controls that ensure the order of cell cycle events. Science 246: 629-634, 1989. Murray, A.W. and Kirshner, M.W. Dominoes and clocks: the union of the views of the cell cycle. Science 246: 614-621, 1989. Murray, A.W. Creative blocks: cell-cycle checkpoints and feed- back controls. Nature 359: 599-604, 1992. Painter, R.B. and Robertson, J.S. Effect of irradiation and theory of role of mitotic delay on the time course of labeling of HeLa S3 cells with tritiated thymidine. Radiat. Res. II: 206-217, 1959. Painter, R.B. The direct effect of X-irradiation on HeLa S3 deoxyribonucleic acid synthesis. Radiat. Res. 16: 846-859, 1962. Yamada, M. and Puck, T.T. Action of radiation on mammalian cells, IV. Reversible mitotic lag in the S3 HeLa cell produced by low doses ofX-rays. Proc. Natl. Acad. Sci. USA 47: 1181-l 191, 1961. Terasima, T. and Tolmach, L.J. Variations in several responses of HeLa cells to X-irradiation during the division cycle. Biophys. J. 3: 11-33, 1963. Yu, C.K. and Sinclair, W.K. Mitotic delay and chromosomal aberrations induced by X-rays in synchronized Chinese hamster cells in vitro. J. Nat. Cancer Inst. 39: 619-632, 1967. Leeper, D.B., Schneiderman, M.H. and Dewey, W.C. Radiation-induced division delay in synchronized Chinese hamster ovary cells in monolayer culture. Radiat. Res. 50: 401-417, 1972. Leeper, D.B., Schneiderman, M.H. and Dewey, WC. Radiation-induced cycle delay in synchronized Chinese hamster cells: comparison between DNA synthesis and division. Radiat. Res. 53: 326-337, 1973. Whitmore, G.F., Stanners, C.P., Till, J.E. and Gulyas, S. Nucle- ic acid synthesis and the division cycle in X-irradiated L-strain mouse cells. B&him. Biophys. Acta 47: 66-77, 1961. Mak, S. and Till, J.E. The effects of X-rays on the progress of L-cells through the cell cycle. Radiat. Res. 20: 600-618, 1963. Lajtha, L.G., Oliver, R., Berry, R. and Noyes, W.D. Mechanism of radiation effect on the process of synthesis of deoxy- ribonucleic acid. Nature 182; 1788-1790, 1958. Lajtha, L.G., Oliver, R., Kumatori, T. and Ellis, F. On the mechanism of radiation effect on DNA synthesis. Radiat. Res. 8: I-16, 1958. Little, J.B. Delayed initiation of DNA synthesis in irradiated human diploid cells. Nature 218: 1064-1065, 1968. Walters, R.A., Gurley, L.R. and Tobey, R.A. Effects of caffeine on radiation-induced phenomena associated with cell-cycle traverse of mammalian cells. Biophys. J. 14: 99- 117, 1974. Tolmach, L.J., Jones, R.W. and Bus%, P.M. The action of caf- feine on X-irradiated HeLa cells. Radiat. Res. 71: 653-665, 1977. Boynton, A.L., Evans, T.C. and Crousc, D.A. Effects of caffeine on radiation-induced mitotic inhibition in S-180 ascites tumor cells. Radiat. Res. 60: 89-97, 1974. McKenna, W.G., Iliakis, G., Weiss, M.C., Bernhard, E.J. and

Page 11: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Maity et al. / Radiother. Oncol. 31 (1994) 1-13

23

24

25

26

27

Muschel, R.J. Increased G2 delay in radiation-resistant cells ob- tained by transformation of primary rat embryo cells with the oncogenes H-ras and V-myc. Radiat. Res. 125: 283-287, 1991. Iliakis, G., Metzher, L., Muschel, R.J. and McKenna, W.G. In- duction and repair of DNA double strand breaks in radiation- resistant cells obtained by transformation of primary rat embryo cells with the oncogenes H-ras and V-myc. Radiat. Res. 50: 6575-6579, 1990. Su, L.N. and Little, J.B. Prolonged cell cycle delay in radioresis- tant human cell lines transfected with activated ras oncogene and/or simian virus 4OT-antigen. Radiat. Res. 133: 73-79, 1993. Norbury, C. and Nurse, P. Animal cell cycles and their control. Annu. Rev. B&hem. 61: 441-470, 1992. Murray, A.W., Solomon, M.J. and Kirschner, M.W. The role of cyclin synthesis and degradation in the control of maturation promoting factor activity. Nature 339: 280-286, 1989. Beach, D., Durkacz, B. and Nurse, P. Functionally homologous cell cycle control genes in budding and fission yeast. Nature 300: 706-709, 1982.

28 Hartwell, L.H., Culotti, J., Pringle, J.R. and Reid, B.J. Genetic control of the cell division cycle in yeast. Science 183: 46-51, 1974.

29

30

31

32 33

Lee. M.G. and Nurse, P. Complementation used to clone a human homologue of the fission yeast cell cycle control gene cdc2. Nature 327: 31-35, 1987. Riabowol, K., Draetta, G., Brizuela, L., Vandre, D. and Beach, D. The cdc2 kinase is a nuclear protein that is essential for mitosis in mammalian cells. Cell 57: 393-401, 1989. Evans, T., Rosenthal, E.T., Youngblom, J., Distel, D. and Hunt, T. Cyclin: a protein specified by maternal mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell 33: 389-396, 1983. Sherr, C.J. Mammalian G, cyclins. Cell 73: 1059-1065, 1993. Booher, R. and Beach, D. Involvement of cdc/3+ in mitotic control in Schizosaccharomyces pombe: possible interaction of the gene product with microtubules. EMBO J. 7: 2321-2327, 1988.

34

35

36

37

38

39

40

41

42

43

Pines, J. and Hunter, T. Isolation of a human cyclin cDNA: evi- dence for cyclin mRNA and protein regulation in the cell cycle and for interaction with ~34’~‘~. Cell 58: 833-846, 1989. Lehner, CF. and O’Farrell, P. Expression and function of Drosophila cyclin A during embryonic cell cycle progression. Cell 56: 957-968, 1989. Hirt, H., Mink, M., Pfosser, M., Bogre, L., Gyorgyey, J., Jonak, C., Gartner, A., Dudits, D. and Heberle-Bors, E. Alfalfa cyclins: differential expression during the cell cycle and in plant organs. Plant Cell 4: 1531-1538, 1992. Markiewicz, D., McKenna, W.G., Flick, M., Maity, A., and Muschel, R.J. The effects of radiation on the expression of a newly cloned and characterized rat cyclin B mRNA. Int. J. Radiat. Oncol., Biol. Phys. 22: 135-144, 1994. Minshull, J., Blow, J.J. and Hunt, T. Translation of cyclin mRNA is necessary for extracts of activated Xenopus eggs to enter mitosis. Cell 56: 947-956, 1989. Richardson, H., Lew, D.J., Henze, M., Sugimoto, K. and Reed, S.1. Cyclin-B homologs in Saccharomyces cerevisiae function in S phase and in Gz. Genes Dev. 6: 2021-2034, 1992. Girard, F., Strausfeld, U., Fernandez, A. and Lamb, N.J.C. Cyclin A is required for the onset of DNA replication in mam- malian fibroblasts. Cell 67: 1169-I 179, 1991. Pagano, M., Pepperkok, R., Verde, F., Ansorge, W. and Draet- ta, G. Cyclin A is required at two points in the human cell cycle. EMBO J. 11: 961-971, 1992. Pines, J. and Hunter, T. Human cyclins A and Bl are differen- tially located in the cell and undergo cell cycle-dependent nu- clear transport. J. Cell. Biol. 115: I-17, 1991. Pines, J. and Hunter, T. Human cyclin A is adenovirus Ela- associated protein p60 and behaves differently from cyclin B. Nature 1990: 760-763, 1990.

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

11

Tsai, L.H., Harlow, E. and Meyerson, M. Isolation of the human c&2 gene that encodes the cyclin A and adenovirus E 1 a associated p33 kinase. Nature 353: 174-178, 1991. Peeper, D.S., Parker, L.L., Ewen, M.E., Toebes, M., Hall, F. L., Xu, M., Zantema, A., Eb, A.J.V. and Piwnica-Worms, H. A and B type cyclins differentially modulate substrate specificity of cyclin cdk complexes. EMBO J. 12: 1947-1954, 1993. Devoto, S.H., Mudryj, M., Pines, J., Hunter, T. and Nevins, J.R. A cyclin A protein kinase complex possesses sequence spe- cific DNA binding activity: ~33“~~ is a component of the E2F cyclin A complex. Cell 68: 167-176, 1992. Gould, K.L. and Nurse, P. Tyrosine phosphorylation of the lis- sion yeast cdc2+ protein kinase regulates entry into mitosis. Nature 342: 39-45, 1989. Gould, K.L., Moreno, S., Owen, D.J., Sazer, S. and Nurse, P. Phosphorylation at Thr167 is required for Schizosaccharomyces pombe ~34’~‘~ function. EMBO J. 10: 3297-3309, 1991. Norbury, C., Blow, J. and Nurse, P. Regulatory phosphoryla- tion of the ~34”~“’ protein kinase in vertebrates. EMBO J. 10: 3321-3329, 1991. Krek, W. and Nigg, E.A. Mutations of ~34~~“~ phosphorylation sites induce premature mitotic events in HeLa cells: evidence for a double block to ~34’~” kinase activation in vertebrates. EMBO J. lo: 3331-3341, 1991. Ducommun, B., Brambilla, P., Felix, M., Franza, J., Karsenti, E. and Draetta, G. cdc2 phosphorylation is required for its interaction with cyclin. EMBO J. 10: 331 I-3319, 1991. Lorca, T., Labbe, J., Devault, A., Fesquet, D.. Capony, J., Cavadore, J., Bouffant, F.L. and Doree, M. Dephosphorylation of cdc2 on threonine 161 is required for cdc2 kinase inactivation and normal anaphase. EMBO J. 11: 2381-2390, 1992. Solomon, M.J., Lee, T. and Kirschner, M.W. Role of phosphor- ylation in ~34~“~ activation: identification of an activating kin- ase. Mol. Biol. Cell 13-27, 1992. Solomon, M.J., Glotzer, M., Lee, T.H., Philippe, M. and Kirschner, M.W. Cyclin activation of ~34”“‘~. Cell 63: 1013-1024, 1990. Atherton-Fessler, S., Parker, L.L., Geahlen, R.L. and Piwnica- Worms, H. Mechanisms of ~34’~“~ regulation. Mol. Cell. Biol. 13: 1675-1685, 1993. Russell, P. and Nurse, P. cdc25 functions as an inducer in the mitotic control of fission yeast. Cell 45: 145-153, 1986. Sadhu, K., Reed, S.I., Richardson, H. and Russell, P. Human homolog of fission yeast cdc25 mitotic inducer is predominantly expressed in GZ. Proc. Natl. Acad. Sci. USA 87: 5139-5143, 1990. Strausfeld, Il., Labbe, J.C., Fesquet, D., Cavadore, J.C., Picard. A., Sadhu, K., Russell, P. and Doree, M. Dephosphorylation and activation of a p34 cd~2/cyclin B complex in vitro by human cdc25 protein. Nature 351: 242-245, 1991. Dunphy, W.G. and Kumagai, A. The cdc25 protein contains an intrinsic phosphatase activity. Cell 67: 189- 196, 199 I. Kumagai, A. and Dunphy, W.G. Regulation of the cdc25 pro- tein during the cell cycle in Xenopus extract. Cell 70: 139-151. 1992. Hoffmann, I., Clarke, P.R., Marcote, M.J., Karsenti, E. and Draetta, G. Phosphorylation and activation of human cdc25-C by cdc2-cyclin B and its involvement in the self-amplification of MPF at mitosis. EMBO J. 12: 53-63, 1993. Russell, P. and Nurse, P. Negative regulation of mitosis by wee1 +, a gene encoding a protein kinase homolog. Cell 49: 559-567, 1987. Featherstone, C. and Russell, P. Fission yeast pl07”‘“r’ mitotic inhibitor is a tyrosine/serine kinase. Nature 349: 808-8 I 1, 199 1. Igarashi, M., Nagata, A., Jinno, S., Suto, K. and Okayama, H. wee]+-like gene in human cells. Nature 353: 80-83. 1991. Parker, L.L. and Piwnica-Worms, H. Inactivation of the

P34 Cdc2_cyc]in B complex by the human wee1 tyrosine kinase. Science 257: 1955-1957, 1992.

Page 12: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

12 A. Muit!, PI ul. / Radiotlrer. Oncol. 31 (1994) I-13

66

61

68

69

70

71

12

13

74

75

76

77

78

79

80

81

82

83

84

McGowan, C.H. and Russell. P. Human wee1 kinase inhibits

cell division by phosphorylating ~34”‘“” exclusively on Tyrl5.

EMBO J. 12: 75-85. 1993. Coleman, T.R., Tang, Z. and Dunphy, W.G. Negative regula-

tion of the wee1 protein kinase by direct action of the niml/cdrl mitotic inducer. Cell 72: 919-929, 1993.

Lundgren, K.. Walworth. N.. Booher. R., Dembski. M..

Kirschner. M. and Beach. D. mik I and weel cooperate in the in-

hibitory tyrosine phosphorylation of cdc2. Cell 64: I I I l-l 122.

1991.

Enoch. T. and Nurse, P. Mutation of fission yeast cell cycle con-

trol genes abolishes dependence of mitosis on DNA replication. Cell 60: 665-673, 1990.

Enoch. T., Carr. A.M. and Nurse, P. Fission yeast genes involv-

ed in coupling mitosis to completion of DNA replication. Genes

Dev. 6: 2035-2046. 1992.

Muschel. R.J.. Zhang. H.B.. lliakis. G. and McKenna. W.G.

Cyclin B expression in HeLa cells during the Gz block induced

by radiation. Cancer Res. 51: 5113-5117. 1991. Muschel. R.J., Zhang. H.B. and McKenna. W.G. Differential

effect of ionizing radiation on the expression of cyclin A and

cyclin B in HeLa cells. Cancer Res. 5: I 128-I 135. 1993.

Datta. R.. Hass, R.. Gunji, H.. Weichselbaum. R. and Kufe. D.

Down-regulation of cell cycle control genes by ionizing radia- tion. Cell Growth Differ. 3: 637-644. 1992.

Lock, R.B. and Ross. W.E. Possible role for ~34”“” kinase in

etoposide-induced cell death of Chinese hamster ovary cells.

Cancer Res. 50: 3767-3771. 1990. Uckun. F.M. Radiation-induced Gz arrest in human cell cycle

is triggered by tyrosine phosphorylation and inactivation of ~34”‘” kinase. In: Forty First Annual Meeting of the Radia-

tion Research Society, 1993 [abstract].

Hain. J., Crompton, N.E.A., Burkart. W. and Jaussi. R. Caf-

feine release of radiation induced S and Gz phase arrest in V79

hamster cells: increase of histone messenger RNA levels and ~34”” -activation. Cancer Res. 53: 1507-1510. 1993. Lock. R.B. Inhibition of ~34““” kinase activation, ~34”““’

tyrosine dephosphorylation. and mitotic progression in Chinese

hamster ovary cells exposed to etoposide. Cancer Res. 52: 1817-1822. 1992.

Tsao, Y.P.. D’Arpa. P. and Liu. L. The involvement of active

DNA synthesis in camptothecin-induced G: arrest: altered regulation of p34 ““J/cyclin B. Cancer Res. 52: 1823-1829.

1992.

O’Connor. P.M.. Ferris, D.K., White. G.A.. Pines, J.. Hunter.

T.. Longo. D.L. and Kohn. K.W. Relationships between cdc2

kinase. DNA cross-linking. and cell cycle perturbations induced

by nitrogen mustard. Cell Growth Differ. 3: 43-52. 1992.

O’Connor. P.M.. Ferris, D.K.. Hoffman. I., Jackman. J.. Draet-

ta. G. and Kohn. K.W. Role of CDC25C phosphatase in Gz ar- rest induced by nitrogen mustard in human lymphoma cells.

Proc. Natl. Acad. Sci. USA (in press).

O’Connor. P.M.. Ferris, D.K., Pagano. M.. Draetta. G.. Pines,

J., Hunter, T.. Longo. D.L. and Kohn, K. Gz delay induced by

nitrogen mustard in human cells affects cyclin A/cdkZ and cyclin

B/cdcZ-kinase complexes differently. J. Biol. Chem. 268: 8298-8308, 1993.

Fingert. H.J.. Pu. A.T.. Chen. 2.. Googe. P.B.. Alley. M. C. and Pardee. A.B. In vivo and in vitro enhanced antitumor effects by pentoxifylline in human cancer cells treated with thiotepa. Cancer Res. 48: 4375-4381. 1988.

Tobey. R.A. Different drugs arrest cells at a number of distinct stages in G,. Nature 254: 245-247. 1975.

Busse. P.M. and Bose, SK. The action of caffeine on X- irradiated HeLa cells. 111. Enhancement of X-ray induced killing

during G? arrest. Radiat. Res. 76: 292-307. 1978.

85 Kim, S.H., Khil. M.S., Ryu, S. and Kim, J.H. Enhancement of

radiation response on human carcinoma cells in culture by pen-

toxifylline. lnt. J. Radiat. Oncol.. Biol. Phys. 25: 61-65. 1992.

86 Weinert. T.A. and Hartwell, L.H. The RAD9 gene controls the

cell cycle response to DNA damage in Saccharomyces cerevisiae. Science 241: 317-322, 1988.

87 Weinert, T.A. and Hartwell, L.H. Characterization of RAD9 of

Saccharomyces cerevisiue and evidence that its function acts

posttranslationally in cell cycle arrest after DNA damage. Mol.

Cell. Biol. IO: 6554-6564. 1990.

88 Weinert. T.A. Dual cell cycle checkpoints sensitive to chromo-

some replication and DNA damage in the budding yeast Sac-

charomyces cerevisiae. Radiat. Res. 132: 141-143, 1992. 89 Rowley. R., Hudson. J. and Young, P.G. The wee1 protein kin-

ase is required for radiation-induced mitotic delay. Nature 356:

353-355. 1992.

90 AI-Khodairy. F. and Carr. A.M. DNA repair mutants defining

Gz checkpoint pathways in Schkosaccharomyces pomhe. EMBO J. II: 1343-1350, 1992.

91 Jimenez. G.. Yucel. J.. Rowley, R. and Subramani. S. The rad3+ gene of S~,hi~osu~~llarom~~es pomhe is involved in multi-

ple checkpoint functions and in DNA repair. Proc. Natl. Acad. Sci. USA 89: 4952-4956. 1992.

92 Rowley. R.. Subramani. S. and Young. P.G. Checkpoint con- trols in Schizosaccharomyces pombe: radl. EMBO J. I I:

1335-1342. 1992.

93 Walworth, N.. Davey, S. and Beach. D. Fission yeast &I pro-

tein kinase links the rad checkpoint pathway to cdc2. Nature 363: 368-370, 1993.

94 Kastan, M.B., Onyekwere. 0.. Sidransky. D.. Vogelstein, B. and

Craig, R.W. Participation of ~53 protein in the cellular response

to DNA damage. Cancer Res. 51: 6304-631 I, 1991.

95 Kuerbitz, S.J.. Plunkett. B.S., Walsh. W.V. and Kastan. M. B.

Wild-type ~53 is a cell cycle checkpoint determinant following irradiation. Proc. Natl. Acad. Sci. USA 89: 7491-7495. 1992.

96 Kastan. M.B.. Zhan. Q.. El-Deiry. W.S.. Carrier. F., Jacks. T., Walsh. W.V.. Plunkett, B.S.. Vogelstein. 8. and Fornace. A.J.

A mammalian cell cycle checkpoint pathway utilizing ~53 and gadd45 is defective in ataxia-telangiectasia. Cell 71: 587-597.

1992.

97 Donehower. L.A.. Harvey. M.. Slagle. B.L.. McArthur. M.J..

Montgomery. C.A.. Jr., Butel, J.S. and Bradley. A. Mice defici- ent for ~53 are developmentally normal but susceptible to spon-

taneous tumours. Nature 356: 215-221. 1992.

98 Fornace. A.J., Nebert, D.W., Hollander, MC.. Leethy. J.D.,

Papathanasiou. M., Fargnoli. J. and Holbrook, N.J. Mammali-

an genes coordinately regulated by growth arrest signals and

DNA-damaging agents. Mol. Cell. Biol. 9: 4196-4203, 1989. 99 Papathanasiou. M.A., Kerr. N.C.K.. Robbins, J.H.. McBride,

O.W.I.. Alamo, J., Barrett. S.F.. Hickson. I.D. and Fornace. A.J. Induction by ionizing radiation of the gadd45 gene in cul-

tured human’cells: lack of mediation by protein kinase C. Mol. Cell. Biol. I I: 1009-1016, 1991.

100 El-Deiry. W.S.. Kern, S.E., Pietenpol. J.A.. Kinzler. K.W. and

Vogelstein, B. Definition of a consensus binding site for ~53.

Nat. Genet. I: 45-49. 1992. 101 O’Connor. P.M., Jackman, J., Jondle. D.. Bhatia, K., Magrath,

I. and I(ohn. K.W. Role of the ~53 tumor suppressor gene in cell cycle arrest and radiosensitivity of Burkitt’s lymphoma cell

lines. Cancer Res. 53: 4776-4780. 1993. 102 Scheffner. M.. Werness. B.A.. Huibregtse. J.M., Levine, A.J.

and Howley. P.M. The E6 oncoprotein encoded by human papillomavirus types16 and I8 promotes the degradation of ~53. Cell 63: I 129-I 136. 1990.

103 Slichenmyer. W.J.. Nelson. W.G.. Slebos. R.J. and Kastan. M.B. LOSS of a ~53 associated G, checkpoint does not decrease

Page 13: The molecular basis for cell cycle delays following ... Advance/journals/radion/ecco37.pdfthe molecular basis for radiosensitivity and decided to use as a model system rat embryo libroblasts

A. Mairy et al. / Radiorher. Oncol. 31 (1994) l-13

104

105

106

107

IO8

109

II0

III

II2

II3

II4

II5 II6

II7

II8

II9

120

I21

122

cell survival following DNA damage. Cancer Res. 53: 4164-4168, 1993. Lee, J.M. and Bernstein, A. ~53 mutations increase resistance to ionizing radiation. Proc. Natl. Acad. Sci. USA 90: 5742-5746. 1993. Lowe, SW.. Schmitt, E.M.. Smith, S.W.. Osborne, B.A. and Jacks, T. ~53 is required for radiation-induced apoptosis in mouse thymocytes. Nature 362: 847-849. 1993. Clarke. A.R.. Purdie. C.A., Harrison, D.J.. Morris. R.G.. Bird. CC.. Hooper. M.L. and Wyllie. A.H. Thymocyte apoptosis in- duced by p53-dependent and independent pathways. Nature 362: 849-852. 1993. Linzer, D.I.H. and Levine, A.J. Characterization of a 54K dalton cellular SV40 tumor antigen present in SV40- transformed cells and uninfected embryonal carcinoma cells. Cell 17: 43-52. 1979. Lane, D.P. and Crawford, L.V. T antigen is bound to a host protein in SV40-transformed cells. Nature 278: 261-263, 1979. Martinez, J., Georgoff, I.. Martinez, J. and Levine, A.J. Cellular localization and cell cycle regulation by a temperature-sensitive ~53 protein. Genes Dev. 5: I5 I - 159, I99 I. Diller. L.. Kassel, J.. Nelson, C.E., Gryka. M.A.. Litwak. G.. Gebhardt. M.. Bressac. 8.. Ozturk. M.. Bake, S.J.. Vogelstein. B. and Friend, S.H. ~53 functions as a cell cycle control protein in osteosarcomas. Mol. Cell. Biol. IO: 5772-5781. 1990. Baker, S.J., Markowitz, S., Fearon, E.R., Willson, J.K.V. and Vogelstein, B. Suppression of human colorectal carcinoma cell growth by wild-type ~53. science 249: 912-914, 1990. Mercer. W.E.. Shields, M.T.. Amin. M., Sauve, G.J.. Appella. E., Romano, J.W. and Ullrich, S.J. Negative growth regulation in a glioblastoma tumor cell line that conditionally expresses human wild-type ~53. Proc. Natl. Acad. Sci. USA 87: 6166-6170. 1990. Chen. P.L., Chen. Y., Bookstein, R. and Lee. W.H. Genetic mechanisms of tumor suppression by the human ~53 gene. Sci- ence 250: 1576-1579. 1990. Hollstein, M., Sidransky. D.. Vogelstein, B. and Harris, C.C. ~53 mutations in human cancers. Science 253: 49-53. 1991. Vogelstein. B. A deadly inheritance. Nature 348: 681-682. 1990. Li, F.P. and Fraumeni, J.F. Soft-tissue sarcomas, breast cancer, and other neoplasms. Ann. Intern. Med. 71: 747-752. 1969. Malkin. D., Li. F.P.. Strong, L.C., Fraumeni. J.. Nelson, C.E.. Kim, D.H., Kassel, J.. Gryka. M.A., Bischoff. F.Z., Tainsky, M.A. and Friend, S.H. Germ line ~53 mutations in a familial syndrome of breast cancer, sarcomas and other neoplasms. Sci- ence 250: 1233-1238. 1990. Fields, S. and Jang, S.K. Presence of a potent transcription acti- vating sequence in the ~53 protein. Science 249: 1046-1048, 1990. Raycroft, L., Wu, H. and Lozano, G. Transcriptional activation by wild-type but not transforming mutants of the ~53 anti- oncogene. Science 249: I O49- IO5 I, 1990. Scharer, E. and Iggo, R. Mammalian ~53 can function as a tran- scription factor in yeast. Nucleic Acids Res. 20: 1539-1545. 1992. Kern, S.E.. Kinzler, K.W.. Bruskin. A., Jarosz. D., Friedman. P., Prives, C. and Vogelstein, B. Identification of ~53 as a sequence-specific DNA-binding protein. Science 252: l708- 1711, 1992. Funk, W.D., Pak, D.T.. Karas. R.H.. Wright, W.E. and Shay. J.W. A transcriptionally active DNA-binding site for human ~53 protein complexes. Mol. Cell. Biol. 12: 2866-2871, 1992.

123

124

I25

126

I27

128

129

130

I31

132 133

134

I35

136

I37

138

139

I40

I41

I3

Kern. SE.. Pietenpol, J.A.. Thiagalingam. S.. Seymour, A., Kinzler, K.W. and Vogelstein. B. Oncogenic forms of ~53 inhi- bit p53-regulated gene expression. Science 256: 827-830, 1992. Farmer, G.. Bargonetti, J.. Zhu, H.. Friedman. P.. Prywes, R. and Prives. C. Wild-type ~53 activates transcription in vitro. Nature 358: 83-85. 1992. Weintraub, H.. Hauschka, S. and Tapscott. S.J. The MCK en- chanter contains a ~53 response element. Proc. Natl. Acad. Sci. USA 88: 4570-4571, 1991. Barak, Y., Juven. T.. Haffner. R. and Oren. M. mdm2 expres- sion is induced by wild type ~53 activity. EMBO J. 12: 461-468, 1993. Ginsberg, D.. Mechta, F., Yaniv, M. and Oren. M. Wild-type ~53 can down-modulate the activity of various promoters. Proc. Natl. Acad. Sci. USA 88: 9979-9983. 1991. Santhanam, U., Ray, A. and Sehgal, P.B. Repression of the interleukin 6 gene promoter by ~53 and the retinoblastoma sus- ceptibility gene product. Proc. Natl. Acad. Sci. USA 88: 7605-7609, 1991. Livingstone. L.R.. White, A.. Sprouse. J.. Livanos. E., Jacks, T. and Tlsty. T.D. Altered cell cycle arrest and gene amplification potential accompany loss of wildtype ~53. Cell 70: 923-935. 1992. Yin, Y.. Tainsky. M.A.. Bischoff. F.Z.. Strong. L.C. and Wahl. G.M. Wild type ~53 restores cell cycle control and inhibits gene amplification in cells with mutant ~53 alleles. Cell 70: 937-948, 1992. Lane. D.P. ~53. guardian of the genome. Nature 358: 15-16. 1992. Lane, D.P. A death in the life of ~53. Nature 362: 786. 1993. Watanabe, 1. Radiation effects on DNA chain growth in mam- malian cells. Radiat. Res. 58: 541-556, 1974. Makino. F. and Okada. S. Effects of ionizing radiation on DNA replication in cultured mammalian cells. Radiat. Res. 62: 37-51, 1975. Painter, R.B. and Young. B.R. X-ray-induced inhibition of DNA synthesis in Chinese hamster ovary, human HeLa. and mouse L cells. Radiat. Res. 64: 648-656, 1975. Painter, R.B. and Young. B.R. Formation of nascent DNA molecules during inhibition of replicon initiation in mammalian cells. B&him. Biophys. Acta 418: 146-I 53. 1976. Walters, R.A. and Hildebrand, C.E. Evidence that X-irradiation inhibits DNA replicon initiation in Chinese hamster cells. B&hem. Biophys. Res. Commun. 65: 265-271, 1975. Povirk. L.F. and Painter, R.B. The effect of 313 nanometer light on initiation of replicons in mammalian cell DNA containing bromodeoxyuridine. B&him. Biophys. Acta 432: 267-272. 1976. Wang, Y. and Iliakis. G. Prolonged inhibition by X-rays of DNA synthesis in cells obtained by transformation of primary rat embryo tibroblasts with oncogenes H-ras and v-myc. Cancer Res. 52: 508-514. 1992. Wang, Y.N.. Cheong, G. and Iliakis. G. Persistent inhibition of DNA synthesis in irradiated rat embryo fibroblasts expressing the oncogenes H-ras plus v-myc derives from inhibition of replicon initiation and is mitigated by staurosporine. Cancer Res. 53: 1213-1217, 1993. Rowley, R. Radiation-induced mitotic delay: a genetic characterization in the fission yeast. Radiat. Res. 132: 144-152. 1992.