Pentoxifylline sensitizes human cervical tumor cells to cisplatin-induced apoptosis by suppressing NF-kappa B and decreased cell senescence

Georgina Hernandez-Flores, Pablo C Ortiz-Lazareno, Jose Manuel Lerma-Diaz, Jorge R Dominguez-Rodriguez, Luis F Jave-Suarez, Adriana del C Aguilar-Lemarroy, Ruth de Celis-Carrillo, Susana del Toro-Arreola, Yessica C Castellanos-Esparza, Alejandro Bravo-Cuellar, Georgina Hernandez-Flores, Pablo C Ortiz-Lazareno, Jose Manuel Lerma-Diaz, Jorge R Dominguez-Rodriguez, Luis F Jave-Suarez, Adriana del C Aguilar-Lemarroy, Ruth de Celis-Carrillo, Susana del Toro-Arreola, Yessica C Castellanos-Esparza, Alejandro Bravo-Cuellar

Abstract

Background: Worldwide, cervical cancer is the second most common causes of cancer in women and represents an important mortality rate. Cisplatin (CIS) is a very important antitumoral agent and can lead tumor cells toward two important cellular states: apoptosis and senescence. In some types of cancers pentoxifylline (PTX) sensitizes these cells to the toxic action of chemotherapeutics drugs such as adriamycin, inducing apoptosis. In the present work, we studied in vitro whether PTX alone or in combination with CIS induces apoptosis and/or senescence in cervix cancer HeLa and SiHa cell lines infected with HPV types 16 and 18, respectively, as well as in immortalized keratinocytyes HaCaT cells.

Methods: HeLa (HPV 18+), SiHa (HPV 16+) cervix cancer cells and non-tumorigenic immortalized HaCaT cells (control) were treated with PTX, CIS or both. The cellular toxicity and survival fraction of PTX and CIS were determinate by WST-1 and clonogenic assays respectively. Apoptosis, caspase activation and phosphorylation of ERK1/2, p38, p65 (NF-κB), Bcl-2 and Bcl-XL anti-apoptotic proteins were determinated by flow cytometry. Senescence by microscopy. Phosphorylation of IκBα and IκB total were measured by ELISA. Pro-apoptotic, anti-apoptotic and senescence genes, as well as HPV-E6/7 mRNA expression, were detected by RT-PCR.

Results: Our results show that after 24 hours of incubation PTX per se is toxic for cancer cells affecting cell viability and inducing apoptosis. The toxicity in HaCaT cells was minimal. CIS induces apoptosis in HeLa and SiHa cells and its effect was significantly increases when the cells were treated with PTX + CIS. In all studies there was a direct correlation with levels of caspases (-3, -6, -7, -9 and -8) activity and apoptosis. CIS induces important levels of senescence and phosphorylation of ERK1/2, p38, p65/RELA, and IκBα, and decreased the expression of anti-apoptotic protein Bcl-XL. Surprisingly these levels were significantly reduced by PTX in tumor cells, and at the same time, increases the expression of pro-apoptotic genes.

Conclusion: PTX sensitizes cervical cancer cells to CIS-induced apoptosis and decreases the CIS-induced senescence in these cells via inhibition of NF-κB signaling pathway; diminishes expression of antiapoptotic proteins and the activation of caspases.

Figures

Figure 1
Figure 1
Determination of late apoptosis and caspase activity of HeLa, SiHa and HaCaT cells after in vitro treatment with pentoxifylline or cisplatin either alone or in combinations. 24 hours later the cells were harvested and late apoptosis was determined by UV light microscopy using ethidium bromide and acridine orange stains, the results represent late apoptosis index (Figure 1A). Caspases-3, -6, - 7, -9 and -8 activation was determined by flow cytometry, the results represent the percentage of caspase activity (Figure 1B and 1C respectively). The results represent the mean ± SD of three independent experiments carried out in triplicate. Statistical analysis, Student's t test. (*) P < 0.001 vs CTL. (♦) P < 0.001 vs CIS.
Figure 2
Figure 2
Determination of β-galactosidase-associated senescence of HeLa, SiHa and HaCaT cells after in vitro treatment with PTX or CIS either alone or in combinations. 24 hours later the cells were harvested and senescence was determined by histochemistry using senescence detection kit (BioVision Mountain View, CA, USA). The results represent the mean ± SD of three independent experiments carried out in triplicate. Statistical analysis, Student's t test. (*) P < 0.001 vs CTL. (♦) P < 0.001 vs CIS.
Figure 3
Figure 3
Phosphorylation of the IκBα [pS32] and IκBα (Total) by ELISA kit of HeLa and SiHa cells after in vitro treatment with pentoxifylline or cisplatin either alone or in combination. 24 hours later the cells were harvested and the phosphorylation of the IκBα [pS32] and IκBα (Total) was determined by commercial ELISA kit (Invitrogen). The results represent the mean ± SD of three independent experiments carried out in triplicate. Statistical analysis Student's t test. (*) P <0.001 vs CTL. (•) P <0.001 vs TNF-α. (♦) P <0.001 vs CIS.
Figure 4
Figure 4
Determination of phosphorylated ERK 1/2, p38, and p65 in HeLa, SiHa and HaCaT cell treatment with pentoxifylline or cisplatin either alone or in combination. 24 hours later the cells were harvested and the phosphorylated ERK1/2, p38 and p65 proteins were determined by flow cytometry. A total of 20,000 events were registered in each test. The results represent the mean ± SD of 3 independent experiments carried out in triplicate. (*) P <0.001 vs untreated control cells. (♦) = P <0.001 vs CIS.
Figure 5
Figure 5
Determination of Bcl-2 and Bcl-XL anti-apoptotic proteins in cervical tumor cells treated with PTX or CIS either alone or in combination. 24 hours later the cells were harvested and the proteins expression were determined by flow cytometry. A total of 20,000 events were registered in each test. The results represent the mean ± SD of 3 independent experiments carried out in triplicate. (*) = P < 0.01 vs CTL. (♦) P < 0.05 vs CIS.
Figure 6
Figure 6
Changes in the expression of caspases, senescence, NF-κB, pro- and antiapoptotic-related genes after in vitro exposure to pentoxifylline or cisplatin either alone or in combination. The gene expressions were determined by real-time quantitative PCR. The data are expressed as mRNA fold-increase using mRNA ribosomal as a reference gene. Experiments were conducted in triplicates and repeated three times. In all cases, SD was not > 0.08.

References

    1. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107.
    1. Boulet GA, Horvath CA, Berghmans S, Bogers J. Human papillomavirus in cervical cancer screening: important role as biomarker. Cancer Epidemiol Biomarkers Prev. 2008;17(4):810–817. doi: 10.1158/1055-9965.EPI-07-2865.
    1. Franco EL, Duarte-Franco E, Ferenczy A. Cervical cancer: epidemiology, prevention and the role of human papillomavirus infection. CMAJ. 2001;164(7):1017–1025.
    1. Hannun YA. Apoptosis and the dilemma of cancer chemotherapy. Blood. 1997;89(6):1845–1853.
    1. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35(4):495–516. doi: 10.1080/01926230701320337.
    1. Herr I, Debatin KM. Cellular stress response and apoptosis in cancer therapy. Blood. 2001;98(9):2603–2614. doi: 10.1182/blood.V98.9.2603.
    1. Ewald JA, Desotelle JA, Wilding G, Jarrard DF. Therapy-induced senescence in cancer. J Natl Cancer Inst. 2010;102(20):1536–1546. doi: 10.1093/jnci/djq364.
    1. Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63(11):2705–2715.
    1. Campisi J. Cellular senescence: putting the paradoxes in perspective. Curr Opin Genet Dev. 2011;21(1):107–112. doi: 10.1016/j.gde.2010.10.005.
    1. Gonzalez VM, Fuertes MA, Alonso C, Perez JM. Is cisplatin-induced cell death always produced by apoptosis? Mol Pharmacol. 2001;59(4):657–663.
    1. Losa JH, Parada Cobo C, Viniegra JG, Sanchez-Arevalo Lobo VJ, Ramon y Cajal S, Sanchez-Prieto R. Role of the p38 MAPK pathway in cisplatin-based therapy. Oncogene. 2003;22(26):3998–4006. doi: 10.1038/sj.onc.1206608.
    1. Lerma-Diaz JM, Hernandez-Flores G, Dominguez-Rodriguez JR, Ortiz-Lazareno PC, Gomez-Contreras P, Cervantes-Munguia R, Scott-Algara D, Aguilar-Lemarroy A, Jave-Suarez LF, Bravo-Cuellar A. In vivo and in vitro sensitization of leukemic cells to adriamycin-induced apoptosis by pentoxifylline. Involvement of caspase cascades and IkappaBalpha phosphorylation. Immunol Lett. 2006;103(2):149–158. doi: 10.1016/j.imlet.2005.10.019.
    1. Gomez-Contreras PC, Hernandez-Flores G, Ortiz-Lazareno PC, Del Toro-Arreola S, Delgado-Rizo V, Lerma-Diaz JM, Barba-Barajas M, Dominguez-Rodriguez JR, Bravo Cuellar A. In vitro induction of apoptosis in U937 cells by perillyl alcohol with sensitization by pentoxifylline: increased BCL-2 and BAX protein expression. Chemotherapy. 2006;52(6):308–315. doi: 10.1159/000096003.
    1. Hernandez-Flores G, Bravo-Cuellar A, Aguilar-Luna JC, Lerma-Diaz JM, Barba-Barajas M, Orbach-Arbouys S. [In vitro induction of apoptosis in acute myelogenous and lymphoblastic leukemia cells by adriamycine is increased by pentoxifylline] Presse Med. 2010;39(12):1330–1331. doi: 10.1016/j.lpm.2010.07.013.
    1. Bravo-Cuellar A, Ortiz-Lazareno PC, Lerma-Diaz JM, Dominguez-Rodriguez JR, Jave-Suarez LF, Aguilar-Lemarroy A, del Toro-Arreola S, de Celis-Carrillo R, Sahagun-Flores JE, de Alba-Garcia JE. et al.Sensitization of cervix cancer cells to Adriamycin by Pentoxifylline induces an increase in apoptosis and decrease senescence. Mol Cancer. 2010;9:114. doi: 10.1186/1476-4598-9-114.
    1. Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–2319. doi: 10.1038/nprot.2006.339.
    1. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70(2):440–446. doi: 10.1158/0008-5472.CAN-09-1947.
    1. Chou TC, Motzer RJ, Tong Y, Bosl GJ. Computerized quantitation of synergism and antagonism of taxol, topotecan, and cisplatin against human teratocarcinoma cell growth: a rational approach to clinical protocol design. J Natl Cancer Inst. 1994;86(20):1517–1524. doi: 10.1093/jnci/86.20.1517.
    1. Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58(3):621–681. doi: 10.1124/pr.58.3.10.
    1. Reynolds CP, Maurer BJ. Evaluating response to antineoplastic drug combinations in tissue culture models. Methods Mol Med. 2005;110:173–183.
    1. Martin D, Lenardo M. Morphological, biochemical, and flow cytometric assays of apoptosis. Curr Protoc Mol Biol. 2001;14 Unit 14 13.
    1. Geske FJ, Lieberman R, Strange R, Gerschenson LE. Early stages of p53-induced apoptosis are reversible. Cell Death Differ. 2001;8(2):182–191. doi: 10.1038/sj.cdd.4400786.
    1. Bohm L, Roos WP, Serafin AM. Inhibition of DNA repair by Pentoxifylline and related methylxanthine derivatives. Toxicology. 2003;193(1-2):153–160. doi: 10.1016/S0300-483X(03)00294-4.
    1. Reddel RR. The role of senescence and immortalization in carcinogenesis. Carcinogenesis. 2000;21(3):477–484. doi: 10.1093/carcin/21.3.477.
    1. Schmitt CA. Cellular senescence and cancer treatment. Biochim Biophys Acta. 2007;1775(1):5–20.
    1. Campisi J, Kim SH, Lim CS, Rubio M. Cellular senescence, cancer and aging: the telomere connection. Exp Gerontol. 2001;36(10):1619–1637. doi: 10.1016/S0531-5565(01)00160-7.
    1. Theron T, Bohm L. Influence of the G2 cell cycle block abrogator pentoxifylline on the expression and subcellular location of cyclin B1 and p34cdc2 in HeLa cervical carcinoma cells. Cell Prolif. 2000;33(1):39–50. doi: 10.1046/j.1365-2184.2000.00160.x.
    1. Theron T, Binder A, Verheye-Dua F, Bohm L. The role of G2-block abrogation, DNA double-strand break repair and apoptosis in the radiosensitization of melanoma and squamous cell carcinoma cell lines by pentoxifylline. Int J Radiat Biol. 2000;76(9):1197–1208. doi: 10.1080/09553000050134438.
    1. Serafin AM, Binder AB, Bohm L. Chemosensitivity of prostatic tumour cell lines under conditions of G2 block abrogation. Urol Res. 2001;29(3):221–227. doi: 10.1007/s002400100186.
    1. Rishi L, Gahlot S, Kathania M, Majumdar S. Pentoxifylline induces apoptosis in vitro in cutaneous T cell lymphoma (HuT-78) and enhances FasL mediated killing by upregulating Fas expression. Biochem Pharmacol. 2009;77(1):30–45. doi: 10.1016/j.bcp.2008.09.018.
    1. Henkels KM, Turchi JJ. Cisplatin-induced apoptosis proceeds by caspase-3-dependent and -independent pathways in cisplatin-resistant and -sensitive human ovarian cancer cell lines. Cancer Res. 1999;59(13):3077–3083.
    1. Horvath B, Vekasi J, Kesmarky G, Toth K. In vitro antioxidant properties of pentoxifylline and vinpocetine in a rheological model. Clin Hemorheol Microcirc. 2008;40(2):165–166.
    1. Bragado P, Armesilla A, Silva A, Porras A. Apoptosis by cisplatin requires p53 mediated p38alpha MAPK activation through ROS generation. Apoptosis. 2007;12(9):1733–1742. doi: 10.1007/s10495-007-0082-8.
    1. Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, Tridandapani S, Anant S, Kuppusamy P. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biol Ther. 2007;6(2):178–184. doi: 10.4161/cbt.6.2.3577.
    1. Yeh PY, Yeh KH, Chuang SE, Song YC, Cheng AL. Suppression of MEK/ERK signaling pathway enhances cisplatin-induced NF-kappaB activation by protein phosphatase 4-mediated NF-kappaB p65 Thr dephosphorylation. J Biol Chem. 2004;279(25):26143–26148. doi: 10.1074/jbc.M402362200.
    1. Zhang Y, Qu X, Jing W, Hu X, Yang X, Hou K, Teng Y, Zhang J, Liu Y. GSTP1 determines cis-platinum cytotoxicity in gastric adenocarcinoma MGC803 cells: regulation by promoter methylation and extracellular regulated kinase signaling. Anticancer Drugs. 2009;20(3):208–214. doi: 10.1097/CAD.0b013e328322fbaa.
    1. Amran D, Sancho P, Fernandez C, Esteban D, Ramos AM, de Blas E, Gomez M, Palacios MA, Aller P. Pharmacological inhibitors of extracellular signal-regulated protein kinases attenuate the apoptotic action of cisplatin in human myeloid leukemia cells via glutathione-independent reduction in intracellular drug accumulation. Biochim Biophys Acta. 2005;1743(3):269–279. doi: 10.1016/j.bbamcr.2004.10.009.
    1. Deng Q, Liao R, Wu BL, Sun P. High intensity ras signaling induces premature senescence by activating p38 pathway in primary human fibroblasts. J Biol Chem. 2004;279(2):1050–1059.
    1. Zubova SG, Bykova TV, Zubova Iu G, Romanov VS, Aksenov ND, Pospelov VA, Pospelova TV. [Role of p38alpha kinase in activation of premature senescence program in transformed mouse fibroblasts] Tsitologiia. 2007;49(2):115–124.
    1. Kwong J, Hong L, Liao R, Deng Q, Han J, Sun P. p38alpha and p38gamma mediate oncogenic ras-induced senescence through differential mechanisms. J Biol Chem. 2009;284(17):11237–11246.
    1. Sethi G, Sung B, Aggarwal BB. Nuclear factor-kappaB activation: from bench to bedside. Exp Biol Med (Maywood) 2008;233(1):21–31. doi: 10.3181/0707-MR-196.
    1. Fiebig AA, Zhu W, Hollerbach C, Leber B, Andrews DW. Bcl-XL is qualitatively different from and ten times more effective than Bcl-2 when expressed in a breast cancer cell line. BMC Cancer. 2006;6:213. doi: 10.1186/1471-2407-6-213.
    1. Suen DF, Norris KL, Youle RJ. Mitochondrial dynamics and apoptosis. Genes Dev. 2008;22(12):1577–1590. doi: 10.1101/gad.1658508.
    1. Chandele A, Prasad V, Jagtap JC, Shukla R, Shastry PR. Upregulation of survivin in G2/M cells and inhibition of caspase 9 activity enhances resistance in staurosporine-induced apoptosis. Neoplasia. 2004;6(1):29–40.
    1. Gazzaniga P, Gradilone A, Petracca A, Nicolazzo C, Raimondi C, Iacovelli R, Naso G, Cortesi E. Molecular markers in circulating tumour cells from metastatic colorectal cancer patients. J Cell Mol Med. 2010;14(8):2073–2077. doi: 10.1111/j.1582-4934.2010.01117.x.
    1. Srivastava RK, Sasaki CY, Hardwick JM, Longo DL. Bcl-2-mediated drug resistance: inhibition of apoptosis by blocking nuclear factor of activated T lymphocytes (NFAT)-induced Fas ligand transcription. J Exp Med. 1999;190(2):253–265. doi: 10.1084/jem.190.2.253.
    1. Shah MA, Schwartz GK. Cell cycle-mediated drug resistance: an emerging concept in cancer therapy. Clin Cancer Res. 2001;7(8):2168–2181.
    1. Yoon SS, Ahn KS, Kim SH, Shim YM, Kim J. In vitro establishment of cis-diammine-dichloroplatinum(II) resistant lung cancer cell line and modulation of apoptotic gene expression as a mechanism of resistant phenotype. Lung Cancer. 2001;33(2-3):221–228. doi: 10.1016/S0169-5002(01)00205-7.
    1. Weller M. Predicting response to cancer chemotherapy: the role of p53. Cell Tissue Res. 1998;292(3):435–445. doi: 10.1007/s004410051072.
    1. Fricker M, Papadia S, Hardingham GE, Tolkovsky AM. Implication of TAp73 in the p53-independent pathway of Puma induction and Puma-dependent apoptosis in primary cortical neurons. J Neurochem. 2010;114(3):772–783. doi: 10.1111/j.1471-4159.2010.06804.x.
    1. Qin JZ, Stennett L, Bacon P, Bodner B, Hendrix MJ, Seftor RE, Seftor EA, Margaryan NV, Pollock PM, Curtis A. et al.p53-independent NOXA induction overcomes apoptotic resistance of malignant melanomas. Mol Cancer Ther. 2004;3(8):895–902.
    1. Perez-Galan P, Roue G, Villamor N, Montserrat E, Campo E, Colomer D. The proteasome inhibitor bortezomib induces apoptosis in mantle-cell lymphoma through generation of ROS and Noxa activation independent of p53 status. Blood. 2006;107(1):257–264. doi: 10.1182/blood-2005-05-2091.
    1. Sankala HM, Hait NC, Paugh SW, Shida D, Lepine S, Elmore LW, Dent P, Milstien S, Spiegel S. Involvement of sphingosine kinase 2 in p53-independent induction of p21 by the chemotherapeutic drug doxorubicin. Cancer Res. 2007;67(21):10466–10474. doi: 10.1158/0008-5472.CAN-07-2090.
    1. Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol. 2011;192(4):547–556. doi: 10.1083/jcb.201009094.
    1. Linskens MH, Harley CB, West MD, Campisi J, Hayflick L. Replicative senescence and cell death. Science. 1995;267(5194):17.

Source: PubMed

3
Abonner