期刊论文详细信息
BMC Medical Genomics
Cidofovir selectivity is based on the different response of normal and cancer cells to DNA damage
Robert Snoeck1  Lieve Naesens1  Dimitri Topalis1  Graciela Andrei1  Tim De Schutter1 
[1] Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, KU Leuven, Leuven, Belgium
关键词: Human papillomavirus;    Gene expression profiling;    DNA damage and repair;    Cidofovir;    Cervical carcinoma;   
Others  :  1092406
DOI  :  10.1186/1755-8794-6-18
 received in 2013-02-01, accepted in 2013-05-17,  发布年份 2013
PDF
【 摘 要 】

Background

Cidofovir (CDV) proved efficacious in treatment of human papillomaviruses (HPVs) hyperplasias. Antiproliferative effects of CDV have been associated with apoptosis induction, S-phase accumulation, and increased levels of tumor suppressor proteins. However, the molecular mechanisms for the selectivity and antitumor activity of CDV against HPV-transformed cells remain unexplained.

Methods

We evaluated CDV drug metabolism and incorporation into cellular DNA, in addition to whole genome gene expression profiling by means of microarrays in two HPV+ cervical carcinoma cells, HPV- immortalized keratinocytes, and normal keratinocytes.

Results

Determination of the metabolism and drug incorporation of CDV into genomic DNA demonstrated a higher rate of drug incorporation in HPV+ tumor cells and immortalized keratinocytes compared to normal keratinocytes. Gene expression profiling clearly showed distinct and specific drug effects in the cell types investigated. Although an effect on inflammatory response was seen in all cell types, different pathways were identified in normal keratinocytes compared to immortalized keratinocytes and HPV+ tumor cells. Notably, Rho GTPase pathways, LXR/RXR pathways, and acute phase response signaling were exclusively activated in immortalized cells. CDV exposed normal keratinocytes displayed activated cell cycle regulation upon DNA damage signaling to allow DNA repair via homologous recombination, resulting in genomic stability and survival. Although CDV induced cell cycle arrest in HPV- immortalized cells, DNA repair was not activated in these cells. In contrast, HPV+ cells lacked cell cycle regulation, leading to genomic instability and eventually apoptosis.

Conclusions

Taken together, our data provide novel insights into the mechanism of action of CDV and its selectivity for HPV-transformed cells. The proposed mechanism suggests that this selectivity is based on the inability of HPV+ cells to respond to DNA damage, rather than on a direct anti-HPV effect. Since cell cycle control is deregulated by the viral oncoproteins E6 and E7 in HPV+ cells, these cells are more susceptible to DNA damage than normal keratinocytes. Our findings underline the therapeutic potential of CDV for HPV-associated malignancies as well as other neoplasias.

【 授权许可】

   
2013 De Schutter et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150128183717275.pdf 3288KB PDF download
Figure 5. 131KB Image download
Figure 4. 213KB Image download
Figure 3. 78KB Image download
Figure 2. 63KB Image download
Figure 1. 127KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

【 参考文献 】
  • [1]Bosch FX, de Sanjosé S, Castellsague X, Moreno V, Muñoz N: Epidemiology of human papillomavirus infection and associations with cervical cancer: new opportunities for prevention. In Papillomavirus Research: From Natural History to Vaccines and Beyond. Edited by Campo MS. UK: Caister Academic Press; 2006:19-40.
  • [2]Doorbar J: Molecular biology of human papillomavirus infection and cervical cancer. Clin Sci (Lond) 2006, 110:525-541.
  • [3]zur Hausen H: Papillomaviruses and cancer: from basic studies to clinical application. Nat Rev Cancer 2002, 2:342-350.
  • [4]Ziegert C, Wentzensen N, Vinokurova S, Kisseljov F, Einenkel J, Hoeckel M, et al.: A comprehensive analysis of HPV integration loci in anogenital lesions combining transcript and genome-based amplification techniques. Oncogene 2003, 22:3977-3984.
  • [5]Baker CC, Phelps WC, Lindgren V, Braun MJ, Gonda MA, Howley PM: Structural and transcriptional analysis of human papillomavirus type 16 sequences in cervical carcinoma cell lines. J Virol 1987, 61:962-971.
  • [6]Bodily J, Laimins LA: Persistence of human papillomavirus infection: keys to malignant progression. Trends Microbiol 2011, 19:33-39.
  • [7]Moody CA, Laimins LA: Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 2010, 10:550-560.
  • [8]Munger K, Basile JR, Duensing S, Eichten A, Gonzalez SL, Grace M, et al.: Biological activities and molecular targets of the human papillomavirus E7 oncoprotein. Oncogene 2001, 20:7888-7898.
  • [9]Howie HL, Katzenellenbogen RA, Galloway DA: Papillomavirus E6 proteins. Virology 2009, 384:324-334.
  • [10]De Clercq E, Holy A: Acyclic nucleoside phosphonates: a key class of antiviral drugs. Nat Rev Drug Discov 2005, 4:928-940.
  • [11]Van Pachterbeke C, Bucella D, Rozenberg S, Manigart Y, Gilles C, Larsimont D, et al.: Topical treatment of CIN 2+ by cidofovir: results of a phase II, double-blind, prospective, placebo-controlled study. Gynecol Oncol 2009, 115:69-74.
  • [12]Snoeck R, Noel JC, Muller C, De Clercq E, Bossens M: Cidofovir, a new approach for the treatment of cervix intraepithelial neoplasia grade III (CIN III). J Med Virol 2000, 60:205-209.
  • [13]Koonsaeng S, Verschraegen C, Freedman R, Bossens M, Kudelka A, Kavanagh J, et al.: Successful treatment of recurrent vulvar intraepithelial neoplasia resistant to interferon and isotretinoin with cidofovir. J Med Virol 2001, 64:195-198.
  • [14]Stier EA, Goldstone SE, Einstein MH, Jay N, Berry JM, Wilkin T, et al.: Safety and efficacy of topical Cidofovir to treat high-grade perianal and vulvar intraepithelial neoplasia in HIV-positive men and women. AIDS 2013, 27:545-551.
  • [15]Tristram A, Fiander A: Clinical responses to Cidofovir applied topically to women with high grade vulval intraepithelial neoplasia. Gynecol Oncol 2005, 99:652-655.
  • [16]Collette DC, Zechel MA: Novel treatment of atypical human papillomavirus-associated epithelial hyperplasia with cidofovir. J Oral Maxillofac Surg 2011, 69:2383-2386.
  • [17]Van Cutsem E, Snoeck R, Van Ranst M, Fiten P, Opdenakker G, Geboes K, et al.: Successful treatment of a squamous papilloma of the hypopharynx-esophagus by local injections of (S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine. J Med Virol 1995, 45:230-235.
  • [18]Andrei G, Snoeck R, Piette J, Delvenne P, De Clercq E: Antiproliferative effects of acyclic nucleoside phosphonates on human papillomavirus (HPV)-harboring cell lines compared with HPV-negative cell lines. Oncol Res 1998, 10:523-531.
  • [19]Andrei G, Snoeck R, Schols D, De Clercq E: Induction of apoptosis by cidofovir in human papillomavirus (HPV)-positive cells. Oncol Res 2000, 12:397-408.
  • [20]Hostetler KY, Rought S, Aldern KA, Trahan J, Beadle JR, Corbeil J: Enhanced antiproliferative effects of alkoxyalkyl esters of cidofovir in human cervical cancer cells in vitro. Mol Cancer Ther 2006, 5:156-159.
  • [21]Abdulkarim B, Sabri S, Deutsch E, Chagraoui H, Maggiorella L, Thierry J, et al.: Antiviral agent Cidofovir restores p53 function and enhances the radiosensitivity in HPV-associated cancers. Oncogene 2002, 21:2334-2346.
  • [22]Sirianni N, Wang J, Ferris RL: Antiviral activity of Cidofovir on a naturally human papillomavirus-16 infected squamous cell carcinoma of the head and neck (SCCHN) cell line improves radiation sensitivity. Oral Oncol 2005, 41:423-428.
  • [23]Amine A, Rivera S, Opolon P, Dekkal M, Biard DS, Bouamar H, et al.: Novel anti-metastatic action of cidofovir mediated by inhibition of E6/E7, CXCR4 and Rho/ROCK signaling in HPV tumor cells. PLoS One 2009, 4:e5018.
  • [24]Johnson JA, Gangemi JD: Selective inhibition of human papillomavirus-induced cell proliferation by (S)-1-[3-hydroxy-2-(phosphonylmethoxy)propyl]cytosine. Antimicrob Agents Chemother 1999, 43:1198-1205.
  • [25]Oliva J, French SW, Li J, Bardag-Gorce F: Proteasome inhibitor treatment reduced fatty acid, triacylglycerol and cholesterol synthesis. Exp Mol Pathol 2012, 93:26-34.
  • [26]Qin S, Chen J, Tanigawa S, Hou DX: Gene expression profiling and pathway network analysis of hepatic metabolic enzymes targeted by baicalein. J Ethnopharmacol 2012, 140:131-140.
  • [27]Andrei G, van den Oord J, Fiten P, Opdenakker G, De Wolf-Peeters C, De Clercq E, et al.: Organotypic epithelial raft cultures as a model for evaluating compounds against alphaherpesviruses. Antimicrob Agents Chemother 2005, 49:4671-4680.
  • [28]Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP: Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 2003, 31:e15.
  • [29]Benjamini Y, Hochberg Y: Controlling the false discovery rate - a practical and powerful approach to multiple testing. J Roy Stat Soc B Met 1995, 57:289-300.
  • [30]Naesens L, Andrei G, Votruba I, Krecmerova M, Holy A, Neyts J, et al.: Intracellular metabolism of the new antiviral compound 1-(S)-[3-hydroxy-2-(phosphonomethoxy)propyl]-5-azacytosine. Biochem Pharmacol 2008, 76:997-1005.
  • [31]Dawson MI, Xia Z: The retinoid X receptors and their ligands. Biochim Biophys Acta 1821, 2012:21-56.
  • [32]DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB: The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 2008, 7:11-20.
  • [33]Chapman JR, Taylor MR, Boulton SJ: Playing the end game: DNA double-strand break repair pathway choice. Mol Cell 2012, 47:497-510.
  • [34]Aziz K, Nowsheen S, Pantelias G, Iliakis G, Gorgoulis VG, Georgakilas AG: Targeting DNA damage and repair: embracing the pharmacological era for successful cancer therapy. Pharmacol Ther 2012, 133:334-350.
  • [35]Halloran PJ, Fenton RG: Irreversible G2-M arrest and cytoskeletal reorganization induced by cytotoxic nucleoside analogues. Cancer Res 1998, 58:3855-3865.
  • [36]Wei SJ, Chao Y, Hung YM, Lin WC, Yang DM, Shih YL, et al.: S- and G2-phase cell cycle arrests and apoptosis induced by ganciclovir in murine melanoma cells transduced with herpes simplex virus thymidine kinase. Exp Cell Res 1998, 241:66-75.
  • [37]Donne AJ, Hampson L, He XT, Rothera MP, Homer JJ, Hampson IN: Cidofovir induces an increase in levels of low-risk and high-risk HPV E6. Head Neck 2009, 31:893-901.
  • [38]Kelley ML, Keiger KE, Lee CJ, Huibregtse JM: The global transcriptional effects of the human papillomavirus E6 protein in cervical carcinoma cell lines are mediated by the E6AP ubiquitin ligase. J Virol 2005, 79:3737-3747.
  • [39]Koromilas AE, Li S, Matlashewski G: Control of interferon signaling in human papillomavirus infection. Cytokine Growth Factor Rev 2001, 12:157-170.
  • [40]Masson M, Hindelang C, Sibler AP, Schwalbach G, Trave G, Weiss E: Preferential nuclear localization of the human papillomavirus type 16 E6 oncoprotein in cervical carcinoma cells. J Gen Virol 2003, 84:2099-2104.
  • [41]Zhang HM, Yuan J, Cheung P, Chau D, Wong BW, McManus BM, et al.: Gamma interferon-inducible protein 10 induces HeLa cell apoptosis through a p53-dependent pathway initiated by suppression of human papillomavirus type 18 E6 and E7 expression. Mol Cell Biol 2005, 25:6247-6258.
  • [42]Henley SA, Dick FA: The retinoblastoma family of proteins and their regulatory functions in the mammalian cell division cycle. Cell Div 2012, 7:10. BioMed Central Full Text
  • [43]Oren M: Regulation of the p53 tumor suppressor protein. J Biol Chem 1999, 274:36031-36034.
  • [44]Moll UM, Petrenko O: The MDM2-p53 interaction. Mol Cancer Res 2003, 1:1001-1008.
  • [45]Perry ME: The regulation of the p53-mediated stress response by MDM2 and MDM4. Cold Spring Harb Perspect Biol 2010, 2:a000968.
  • [46]Real PJ, Sanz C, Gutierrez O, Pipaon C, Zubiaga AM, Fernandez-Luna JL: Transcriptional activation of the proapoptotic bik gene by E2F proteins in cancer cells. FEBS Lett 2006, 580:5905-5909.
  • [47]Chinnadurai G, Vijayalingam S, Rashmi R: BIK, the founding member of the BH3-only family proteins: mechanisms of cell death and role in cancer and pathogenic processes. Oncogene 2008, 27(Suppl 1):S20-S29.
  • [48]Deng LL, Shao YX, Lv HF, Deng HB, Lv FZ: Over-expressing CYLD augments antitumor activity of TRAIL by inhibiting the NF-kappaB survival signaling in lung cancer cells. Neoplasma 2012, 59:18-29.
  • [49]Yang ZR, Dong WG, Lei XF, Liu M, Liu QS: Overexpression of Dickkopf-3 induces apoptosis through mitochondrial pathway in human colon cancer. World J Gastroenterol 2012, 18:1590-1601.
  • [50]Yang H, Choi HJ, Park SH, Kim JS, Moon Y: Macrophage inhibitory cytokine-1 (MIC-1) and subsequent urokinase-type plasminogen activator mediate cell death responses by ribotoxic anisomycin in HCT-116 colon cancer cells. Biochem Pharmacol 2009, 78:1205-1213.
  • [51]Kallio JP, Hopkins-Donaldson S, Baker AH, Kahari VM: TIMP-3 promotes apoptosis in nonadherent small cell lung carcinoma cells lacking functional death receptor pathway. Int J Cancer 2011, 128:991-996.
  • [52]Ren C, Li L, Goltsov AA, Timme TL, Tahir SA, Wang J, et al.: mRTVP-1, a novel p53 target gene with proapoptotic activities. Mol Cell Biol 2002, 22:3345-3357.
  • [53]Li L, Ren C, Yang G, Fattah EA, Goltsov AA, Kim SM, et al.: GLIPR1 suppresses prostate cancer development through targeted oncoprotein destruction. Cancer Res 2011, 71:7694-7704.
  • [54]Reza HM, Nishi H, Kataoka K, Takahashi Y, Yasuda K: L-Maf regulates p27kip1 expression during chick lens fiber differentiation. Differentiation 2007, 75:737-744.
  • [55]Hegde SP, Zhao J, Ashmun RA, Shapiro LH: c-Maf induces monocytic differentiation and apoptosis in bipotent myeloid progenitors. Blood 1999, 94:1578-1589.
  • [56]Peng S, Lalani S, Leavenworth JW, Ho IC, Pauza ME: c-Maf interacts with c-Myb to down-regulate Bcl-2 expression and increase apoptosis in peripheral CD4 cells. Eur J Immunol 2007, 37:2868-2880.
  • [57]Hale TK, Myers C, Maitra R, Kolzau T, Nishizawa M, Braithwaite AW: Maf transcriptionally activates the mouse p53 promoter and causes a p53-dependent cell death. J Biol Chem 2000, 275:17991-17999.
  • [58]Gualdrini F, Corvetta D, Cantilena S, Chayka O, Tanno B, Raschella G, et al.: Addiction of MYCN amplified tumours to B-MYB underscores a reciprocal regulatory loop. Oncotarget 2010, 1:278-288.
  • [59]Sottile F, Gnemmi I, Cantilena S, D’Acunto WC, Sala A: A chemical screen identifies the chemotherapeutic drug topotecan as a specific inhibitor of the B-MYB/MYCN axis in neuroblastoma. Oncotarget 2012, 3:535-545.
  • [60]Lu X, Pearson A, Lunec J: The MYCN oncoprotein as a drug development target. Cancer Lett 2003, 197:125-130.
  • [61]Bonauer A, Dimmeler S: The microRNA-17-92 cluster: still a miRacle? Cell Cycle 2009, 8:3866-3873.
  • [62]De Brouwer S, Mestdagh P, Lambertz I, Pattyn F, De PA, Westermann F, et al.: Dickkopf-3 is regulated by the MYCN-induced miR-17-92 cluster in neuroblastoma. Int J Cancer 2012, 130:2591-2598.
  • [63]Frenzel A, Zirath H, Vita M, Albihn A, Henriksson MA: Identification of cytotoxic drugs that selectively target tumor cells with MYC overexpression. PLoS One 2011, 6:e27988.
  • [64]Gabay C, Kushner I: Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 1999, 340:448-454.
  • [65]Sperka T, Wang J, Rudolph KL: DNA damage checkpoints in stem cells, ageing and cancer. Nat Rev Mol Cell Biol 2012, 13:579-590.
  • [66]Jakobsson T, Treuter E, Gustafsson JA, Steffensen KR: Liver X receptor biology and pharmacology: new pathways, challenges and opportunities. Trends Pharmacol Sci 2012, 33:394-404.
  • [67]El Roz A, Bard JM, Huvelin JM, Nazih H: LXR agonists and ABCG1-dependent cholesterol efflux in MCF-7 breast cancer cells: relation to proliferation and apoptosis. Anticancer Res 2012, 32:3007-3013.
  • [68]Rathinam R, Berrier A, Alahari SK: Role of Rho GTPases and their regulators in cancer progression. Front Biosci 2011, 16:2561-2571.
  • [69]Zhang B, Zhang Y, Dagher MC, Shacter E: Rho GDP dissociation inhibitor protects cancer cells against drug-induced apoptosis. Cancer Res 2005, 65:6054-6062.
  • [70]Lazer G, Katzav S: Guanine nucleotide exchange factors for RhoGTPases: good therapeutic targets for cancer therapy? Cell Signal 2011, 23:969-979.
  文献评价指标  
  下载次数:18次 浏览次数:5次