search
for
 About Bioline  All Journals  Testimonials  Membership  News


Journal of Cancer Research and Therapeutics
Medknow Publications on behalf of the Association of Radiation Oncologists of India (AROI)
ISSN: 0973-1482 EISSN: 1998-4138
Vol. 1, Num. 3, 2005, pp. 147-150

Journal of Cancer Research and Therapeutics, Vol. 1, No. 3, July-September, 2005, pp. 147-150

Original Article

The role of the p53 molecule in cancer therapies with radiation and/or hyperthermia.

Department of Biology, Nara Medical University School of Medicine, Kashihara, Nara 634-8521, Japan

Correspondence Address: Ohnishi T, Department of Biology, Nara Medical University School of Medicine, Shijo-cho 840, Kashihara, Nara 6348521, Japan. E-mail: tohnishi@naramedu.ac.jp

Code Number: cr05034

Abstract

In recent years, cancer-related genes have been analyzed as predictive indicators for cancer therapies. Among those genes, the gene product of a tumor suppressor gene p53 plays an important role in cancer therapy, because the p53 molecule induces cell-cycle arrest, apoptosis and depression of DNA repair after cancer therapies such as radiation, hyperthermia and anti-cancer agents. An abnormality of the p53 gene might introduce low efficiency in their cancer therapies. Mutations of p53 are observed at a high frequency in human tumors, and are recognized in about half of all malignant tumors in human. In the both systems of a human cell culture and their transplanted tumor, the sensitivities to radiation, heat and anti-cancer agents were observed in wild-type p53 cells, but not in mutated or deleted p53 cells. In this review, we discuss the p53 activation signaling pathways through the modification of p53 molecules such as phosphorylation after radiation and/or hyperthermia treatments.

Keywords: p53, Predictive indicator, Cancer therapy, X-ray, Hyperthermia, Apoptosis

INTRODUCTION

Combinations of radiation and hyperthermia therapies have been widely adopted for interdisciplinary cancer therapy. It is well known that ionizing radiation-induced cell killing is enhanced by hyperthermia in vitro .[1],[2] Previous studies have shown that heat treatment depresses the DNA repair of radiation-induced DNA strand breaks and thymine lesions.[3],[4] In addition, it has been reported that the activities of DNA polymerases, α and β, are sensitive to heat treatment at temperatures higher than 40°C.[5],[6] Thus, it has been understood that the synergistic effects of hyperthermia on radiation-induced cell killing is induced mainly through the inhibition of DNA repair mechanisms. Moreover, another possible mechanism of radiosensitization by hyperthermia has been suggested to involve the hyperthermic instability of the Ku subunits of DNA-PK, which contribute to the repair of radiation-induced double-strand breaks in DNA.[7], [8]

Patients with tumors that have p53 mutations often have a worse prognosis than those with tumors that don′t have wild-type p53 (wtp53).[9] For prognosis-predictive assays of cancer therapy, the genetic status of the p53 gene is the most important candidate among various cancer-related genes.[10] We previously reported that the radio-, heat- and chemo-sensitivities of human cultured tongue squamous cell carcinoma cells are p53-dependent, and are closely correlated with the induction of apoptosis in in vitro- 11],[12],[13] and in vivo . [14],[15],[16] The interactive hyperthermic enhancement of radiosensitivity was also observed in wtp53 cells, but not in mutated p53 (mp53) cells.[17] However, it remains unclear whether the hyperthermic enhancement of tumor growth inhibition by irradiation is p53-dependent. To clarify the problem, we described in this review whether the p53 gene products contribute to the hyperthermic enhancement of tumor growth inhibition by X-ray irradiation, using transplantable human cultured tongue squamous cell carcinoma cells with an identical genotype except for the p53 gene status, as an in vivo experimental model from the view point of p53 activation and deactivation through the modification and degradation of p53 molecules.

p53 and the Signal Pathways

The ancestor of the mammalian p53 tumor suppressor protein is homologous to drosophila and nematodes.[18] The gene product prevents the malignant degeneration of a normal cell. In cancer cells bearing m p53 , genetic stability is lost and mutations are accumulated, and therefore, malignant changes in the cancer progress at a high frequency. p53 -mutant and p53 -deleted cells account for about 50% of total advanced cancer cells.[19] The p53 molecules are modified for activation by many kinds of different protein kinases at different portions after cell stress [Figure - 1].[20]

p53 regulates the transcription of the phenotypic expressions of target genes by binding to a specific sequence. One of the p53 target genes, WAF1 (wild-type p53 activated fragment 1) inactivates PCNA regulating DNA replication,[21] and induces p53 -dependent G1 arrest through the inhibition of cyclin/CDK activity.[22], [23] During cell cycle arrest, p53 -regulated pathways, such as gadd45 (growth arrest and DNA damage inducible 45) and p53R2 (ribonucleotide reductase small subunit 2), play an important role in the repair of damaged DNA.[24], [25] Otherwise, DNA damage induces apoptosis by several processes of p53 -regulated pathways, such as Bax (Bcl-associated X protein),[26] Fas/APO-1 ,[27] and PAG608 .[28] In contrast, p53 -regulated MDM 2 (murine double minute 2)[29] functions in the negative feedback regulation of p53 activity.[26] It is reported that these determinations of cell cycle arrest or apoptosis are divided by the modification of p53 molecules, such as phosphorylation [Figure - 1],[30] acetylation,[31] poly (ADP-ribosyl)ation,[32] and sumoylation[33],[34]. When the cells are stressed, Ser15/Ser20 of p53 is phosphorylated, then MDM 2 is separated from the phosphorylated p53, and finally p53 is stabilized and activated. [35],[36],[37] Thereafter, p53 binds to the promoter of WAF1 or the p53R2 gene related to DNA repair, and induces their gene expression. When there are too many DNA lesions, however, G1 arrest and DNA repair do not succeed. In this case, p53 is phosphorylated at Ser46, and then binds to the promoter of the p53AIP1 (p53-regulated Apoptosis- Inducing Protein 1) gene. Therefore, p53AIP1 is accumulated. Apoptosis happens in the damaged cells.[38],[39] Other p53 modifications of acetylation and sumoylation have also been reported. It is understood that acetylation is performed at the C-terminal region of p53, and the binding capacity of p53 to specific DNA is enhanced.[40] In addition, there are several reports regarding the sumoylation of lysine 386 in p53, which seems to activate a p53 -downstream transcriptional factor.[33], [34],[41]-[43] In these modifications, it is thought that the structural change of p53 molecules, which bind to specific DNA sequences known as p53 CON, is brought about by many kinds of the up-stream genes. On the other hand, p53 molecules are deactivated and degradated by activated MDM2 molecules, which are phosphorylated at multi-sites by other protein kinases [Figure - 1].

Moreover, p53 is reported to bind to other proteins, such as heat shock proteins (HSPs), which are a famous stress protein.[44], [45] Thus, p53 regulates the fate of the cells after stresses, such as cancer therapies.

p53 Dependent Synergism after Hyperthermia and X-ray Treatment

We previously reported that radio- and heat-sensitivities of cultured human tongue squamous cell carcinoma cells are p53 -dependent, and are closely correlated with the induction of apoptosis in vitro .[11], [12] To confirm that the hyperthermic enhancement of tumor growth inhibition by X-ray irradiation is dependent on the p53 gene status, we used two kinds of cancer cell lines carrying a different p53 gene status, wt p53 and m p53 . We compared heat and X-ray induced cell killing and apoptosis frequencies in the wt p53 cells and the m p53 cells though Bax and Caspase-3 pathways.[11], [12] We adopted mild treatments with radiation and hyperthermia for transplantation on nude mice to examine the synergistic effects on tumor growth inhibition. Tumor growth curves are shown in [Figure - 2]A.

We applied an individual treatment with hyperthermia at 42°C for 20 min or X-irradiation (2 Gy) which showed almost no effect on the growth curves of the both wt p53 and m p53 tumors in the non-treated control groups [Figure - 2]A. When the tumors were treated with a combination of X-rays and hyperthermia, the apparent enhancement of tumor growth inhibition was observed in the wt p53 tumors, but not the m p53 tumors.

We examined both the accumulation of apoptosis-related proteins and the incidence of apoptosis by immunohistochemical analysis [Figure - 2]B. The apoptosis incidence was apparently higher in the wt p53 tumors 72 h after the combined treatment, but not in the m p53 tumors. The fragmentation of PARP and Caspase-3 as the activation of Caspase-3 showed almost the same pattern as the incidence of apoptosis in the wt p53 tumor.[46]

p53 Gene Status as a Predictive Indicator in Cancer Therapy

As previously reported, if the hyperthermic enhancement of radiosensitivity results in heat-induced denaturation of repair enzymes for DNA damage, the synergism of the combination therapy of radiation and hyperthermia should be similarly detected in both wt p53 and m p53 tumors. However, it is very interesting that the synergistic depression of tumor growth was found only in the wt p53 tumors[46]). These findings suggest that the hyperthermic enhancement of tumor growth inhibition by irradiation may result in p53 -dependent apoptosis due to heat-induced inactivation of the cell survival system (s), through either regulation of the cell cycle or induction of DNA repair. Thus, m p53 tumors may rarely include this function, which promotes cell killing by the heat-induced interactive inactivation of the repair enzyme (s) for certain types of sublethal damage. As participants in the p53 -dependent apoptotic processes, the induction of apoptosis mediated by caspase-3 activation was examined in these tumors. In fact, activated Caspase-3 was confirmed from the proteolysis of several important molecules, such as PARP and Caspase-3 itself so called markers of apoptosis.[47],[48] The induction of p53 -dependent apoptosis showed a pattern similar to the tumor growth inhibition after combined treatments with radiation and hyperthermia [Figure - 3]. Therefore, gene analysis of the p53 gene status of cancer cells can be used as a predictive assay for the effectiveness of combined therapy with radiation and hyperthermia [Figure - 3].

ACKNOWLEDGMENT

This work was supported by Grants-in-Aid from the Ministry of Education, Science, Sports and Culture of Japan.

References

1.Belli J, Bonte F. Influence of temperature on the radiation response of mammalian cells in tissue culture. Radiat Res 1963;18:272-6.  Back to cited text no. 1    
2.Ben-Hur E, Elkind MM, Bronk BV. Thermally enhanced radioresponse of cultured Chinese hamster cells: inhibition of repair of sublethal damage and enhancement of lethal damage. Radiat Res 1974;58:38-51.  Back to cited text no. 2  [PUBMED]  
3.Clark EP, Dewey WC, Lett JT. Recovery of CHO cells from hyperthermic potentiation to X-rays repair of DNA and chromatin. Radiat Res 1981;85:302-13.  Back to cited text no. 3  [PUBMED]  
4.Warters RL, Roti Roti JL. Excision of X-ray-induced thymine damage in chromatin from heated cells. Radiat Res 1979;79:113-21.  Back to cited text no. 4  [PUBMED]  
5.Raaphorst GP, Feeley MM, Chu GL, Dewey WC. A comparison of the enhancement of radiation sensitivity and DNA polymerase inactivation by hyperthermia in human glioma cells. Radiat Res 1993;134:331-6.  Back to cited text no. 5  [PUBMED]  
6.Dube DK, Seal G, Loeb LA. Differential heat sensitivity of mammalian DNA polymerases. Biochem Biophys Res Comm 1976;76:483-7.  Back to cited text no. 6  [PUBMED]  
7.Matsumoto Y, Suzuki N, Sakai K, Morimatsu A, Hirano K, Murofushi H. A possible mechanism for hyperthermic radiosensitization mediated through hyperthermic lability of Ku subunits in DNA-dependent protein kinase. Biochem Biophys Res Comm 1997;234:568-72.  Back to cited text no. 7  [PUBMED]  [FULLTEXT]
8.Burgman P, Ouyang H, Peterson S, Chen D J, Li GC. Heat inactivation of Ku autoantigen: possible role in hyperthermic radiosensitization. Cancer Res 1997;57:2847-50.  Back to cited text no. 8    
9.Lowe SW. Cancer therapy and p53. Curr. Opin. Oncol 1995;7:547-53.  Back to cited text no. 9  [PUBMED]  
10.Velculescu VE, El-Deiry WS. Biological and clinical importance of the p53 tumor suppressor gene. Clin Chem 1996;42:858-68.  Back to cited text no. 10  [PUBMED]  
11. Ota I, Ohnishi K, Takahashi A, Yane K, Kanata H, Miyahara H, et al . Transfection with mutant p53 gene inhibits heat-induced apoptosis in a head and neck cell line of human squamous cell carcinoma. Int J Radiat Oncol Biol Phys 2000;47:495-501.  Back to cited text no. 11
12.Takahashi A. Different inducibility of radiation- or heat-induced p53 -dependent apoptosis after acute or chronic irradiation in human cultured squamous cell carcinoma cells. Int J Radiat Biol 2001;77:215-24.  Back to cited text no. 12    
13.Ohnishi K, Ota I, Takahashi A, Yane K, Ohnishi T. Transfection of mutant p53 gene depresses X-ray- or CDDP-induced apoptosis in a human squamous cell carcinoma of the head and neck. Apoptosis 2002;7:367-72.  Back to cited text no. 13    
14.Asakawa I, Yoshimura H, Takahashi A, Ohnishi K, Nakagawa H, Ota I, et al . Growth inhibition in transplanted human tongue tumors bearing different p53 gene status with X-ray or C-beam irradiation. Anticancer Res 2002;22:2037-43.  Back to cited text no. 14    
15.Tamamoto T, Yoshimura H, Takahashi A, Asakawa I, Ota I, Nakagawa H, et al . Heat-induced growth inhibition and apoptosis in transplanted human head and neck squamous cell carcinomas with different p53 status. Int J Hyperthermia 2003;19:590-7.  Back to cited text no. 15    
16.Yuki K, Takahashi A, Ota I, Ohnishi K, Yasumoto J, Yane K, et al . ensitization by glycerol for CDDP-therapy against human cultured cancer cells and tumors bearing mutated p53 gene. Apoptosis 2004;9:853-9.  Back to cited text no. 16    
17.Takahashi A, Ohnishi K, Ota I, Asakawa I, Tamamoto T, Furusawa Y, et al . p53-dependent thermal enhancement of cellular sensitivity in human squamous cell carcinomas in relation to LET. Int J Radiat Biol 2001;77:1043-51.   Back to cited text no. 17    
18.Derry WB, Putzke AP, Rothman JH. Caenorhabditis elegans p53: role in apoptosis, meiosis, and stress resistance. Science 2001;294:591-5.  Back to cited text no. 18    
19. Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutation in human cancers. Science 1991;253:49-53.  Back to cited text no. 19
20.Wang X, Ohnishi T. p53-dependent signal transduction induced by stress. J Radiat Res 1997;38:179-94.  Back to cited text no. 20    
21.Bambara RA, Jessee CB. Properties of DNA polymerases delta and epsilon, and their roles in eukaryotic DNA replication. Biochim Biophys Acta 1991;1088:11-24.  Back to cited text no. 21    
22.El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, et al . WAF1, a potential mediator of p53 tumor suppression. Cell 1993;75:817-25.  Back to cited text no. 22    
23.Deng C, Zhang P, Harper JW, Elledge SJ, Leder P. Mice lacking p21CIP1/WAF1 undergo normal development, but are defective in G1 checkpoint control. Cell 1995;82:675-84.  Back to cited text no. 23    
24.Kastan MB, Zhan Q, El-Deiry WS, Carrier F, Jacks T, Walsh WV, et al . A mammalian cell cycle checkpoint pathway utilizing p53 and GADD45 is defective in ataxia-telangiectasia. Cell 1992;71:587-97.  Back to cited text no. 24    
25.Smith ML, Kontny HU, Zhan Q, Sreenath A, O'Connor PM, Fornace A J Jr. Antisense GADD45 expression results in decreased DNA repair and sensitizes cells to u.v.-irradiation or cisplatin. Oncogene 1996;13:2255-63.  Back to cited text no. 25    
26. Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 1995;80:293-9.  Back to cited text no. 26    
27.Owen-Schaub LB, Zhang W, Cusack JC, Angelo LS, Santee SM, Fujiwara T, et al . Wild-type human p53 and a temperature-sensitive mutant induce Fas/APO-1 expression. Mol Cell Biol 1995;15:3032-40.  Back to cited text no. 27    
28.Israeli D, Tessler E, Haupt Y, Elkeles A, Wilder S, Amson R, et al . A novel p53-inducible gene, PAG608, encodes a nuclear zinc finger protein whose overexpression promotes apoptosis. EMBO J 1997;16:4384-92.  Back to cited text no. 28    
29. Haupt S, Louria-Hayon I, Haupt Y. P53 licensed to kill? Operating the assassin. J Cell Biochem 2003;88:76-82. Back to cited text no.9
30.Barak Y, Oren M. Enhanced binding of a 95 kDa protein to p53 in cells undergoing p53-mediated growth arrest. EMBO J 1992;11:2115-21.  Back to cited text no. 30    
31.Brooks CL, Gu W. Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr Opin Cell Biol 2003;5:164-71.  Back to cited text no. 31    
32.Valenzuela MT, Guerrero R, Nunez MI, Ruiz De Almodovar JM, Sarker M, de Murcia G, et al . PARP-1 modifies the effectiveness of p53-mediated DNA damage response. Oncogene 2002;21:1108-16.  Back to cited text no. 32    
33.Schmidt D, Muller S. Members of the PIAS family act as SUMO ligases for c-Jun and p53 and repress p53 activity. Proc Natl Acad Sci USA 2001;99:2872-7.  Back to cited text no. 33    
34.Melchior F, Hengst L. SUMO-1 and p53. Cell Cycle 2002;1:245-9.  Back to cited text no. 34    
35.Siliciano JD, Canman CE, Taya Y, Sakaguchi K, Appella E, Kastan MB. DNA damage induces phosphorylation of the amino terminus of p53. Genes Dev 1997;11:3471-81.  Back to cited text no. 35    
36.Shieh SY, Ahn J, Tamai K, Taya Y, Prives C. The human homologs of checkpoint kinases Chk1 and Cds1 (Chk2) phosphorylate p53 at multiple DNA damage-inducible sites. Genes Dev 2000;14:289-300.  Back to cited text no. 36    
37.Urban G, Golden T, Aragon IV, Cowsert L, Cooper SR, Dean NM, et al . Identification of a functional link for the p53 tumor suppressor protein in dexamethasone-induced growth suppression. J Biol Chem 2003;278:9747-53.  Back to cited text no. 37    
38.Oda K, Arakawa H, Tanaka T, Matsuda K, Tanikawa C, Mori T, et al . p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell 2000;102:849-62.  Back to cited text no. 38    
39.Saito S, Goodarzi AA, Higashimoto Y, Noda Y, Lees-Miller SP, Appella E, et al . ATM mediates phosphorylation at multiple p53 sites, including Ser (46), in response to ionizing radiation. J Biol Chem 2002;277:12491-4.  Back to cited text no. 39    
40.Gu W, Roeder RG. Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 1997;90:595-606.  Back to cited text no. 40    
41.Zhong S, Muller S, Ronchetti S, Freemont PS, Dejean A, Pandolfi PP. Role of SUMO-1-modified PML in nuclear body formation. Blood 2000;95:2748-52.  Back to cited text no. 41    
42.Rodriguez MS, Desterro JM, Lain S, Midgley CA, Lane DP, Hay RT. SUMO-1 modification activates the transcriptional response of p53. EMBO J 1999;18:6455-61.  Back to cited text no. 42    
43.Gostissa M, Hengstermann A, Fogal V, Sandy P, Schwarz SE, Scheffner M, et al . Activation of p53 by conjugation to the ubiquitin-like protein SUMO-1. EMBO J 1999;18:6462-71.  Back to cited text no. 43    
44.Ohnishi T, Matsumoto H, Takahashi A, Shimura M, Majima H. Accumulation of mutant p53 and hsp72 by heat treatment, and their association in a human glioblastoma cell line. Int J Hyperthermia 1995;11:663-71.  Back to cited text no. 44    
45.Matsumoto H, Wang X, Ohnishi T. Binding between wild-type p53 and hsp72 accumulated after UV and gamma-ray irradiation. Cancer Lett 1995;92:127-33.  Back to cited text no. 45    
46.Takahashi A, Ota I, Tamamoto T, Asakawa I, Nagata Y, Nakagawa H, et al . p53-dependent hyperthermic enhancement of tumour growth inhibition by X-ray or carbon-ion beam irradiation. Int J Hyperthermia 2003;19:145-53.  Back to cited text no. 46    
47.Streffer C, van Beuningen D. The biological basis for tumour therapy by hyperthermia and radiation. Recent Results Cancer Res 1987;104, 24-70.  Back to cited text no. 47    
48.Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG. Specific proteolytic cleavage of poly (ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res 1993; 53:3976-85.  Back to cited text no. 48    

Copyright 2005 - Journal of Cancer Research and Therapeutics


The following images related to this document are available:

Photo images

[cr05034f3.jpg] [cr05034f1.jpg] [cr05034f2.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil