全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

DTNQ-Pro, a Mimetic Dipeptide, Sensitizes Human Colon Cancer Cells to 5-Fluorouracil Treatment

DOI: 10.1155/2013/509056

Full-Text   Cite this paper   Add to My Lib

Abstract:

The resistance of growing human colon cancer cells to chemotherapy agents has been correlated to endogenous overexpression of stress proteins including the family of heat shock proteins (HSPs). Previously, we have demonstrated that a quinone-based mimetic dipeptide, named DTNQ-Pro, induced differentiation of growing Caco-2 cells through inhibition of HSP70 and HSP90. In addition, our product induced a HSP27 and vimentin intracellular redistribution. In the present study, we have evaluated whether a decrease of stress proteins induced by DTNQ-Pro in Caco-2 cells could sensitize these cells to treatment with 5-fluorouracil (5-FU) cytotoxicity. The pretreatment of Caco-2 with 500?nM of DTNQ-Pro increases lipid peroxidation and decreases expression of p38 mitogen-activated protein kinase (MAPK) and FOXO3a. At the same experimental conditions, an increase of the 5-FU-induced growth inhibition of Caco-2 cells was recorded. These effects could be due to enhanced DTNQ-Pro-induced membrane lipid peroxidation that, in turn, causes the sensitization of cancer cells to the cytotoxicity mediated by 5-FU. 1. Introduction Adenocarcinoma cells, such as colorectal cancer (CRC) cells, are remarkably resistant to radiation or chemotherapy-induced damage. As a consequence, the tumours are hard to treat and often proliferate rapidly, even under conditions that may adversely affect normal cells. For several years, 5-fluorouracil (5-FU), a pyrimidine antimetabolite, has been the drug of choice for the treatment of CRC as well as head and neck, pancreatic, and breast carcinomas. 5-FU is known to block DNA synthesis by the inhibition of thymidylate synthase (TS), which is regulated by cell cycle proteins controlled by phosphorylation [1]. Unfortunately, many of the schedules based upon 5-FU alone or in combination with other agents become ineffective during the course of the treatment due to the occurrence of drug resistance to 5-FU. Between several survival pathways activated in cancer cells to antagonize the antiproliferative activities of antineoplastic agents [2–4]. The mechanisms underlying the survival advantage can also be partially related to the increased expression of stress proteins [5, 6]. In fact, in contrast to normal cells, the basal levels of inducible heat shock proteins (HSPs) are frequently higher in tumour cells [7, 8]. The high expression of members of the HSP family in CRC cells has been associated with both metastases and resistance to chemotherapy. Moreover, in experimental models, HSP27 and HSP70 have been shown to increase tumorigenicity of cancer

References

[1]  J. G. Kuhn, “Fluorouracil and the new oral fluorinated pyrimidines,” Annals of Pharmacotherapy, vol. 35, no. 2, pp. 217–227, 2001.
[2]  G. Vitale, C. H. J. van Eijck, P. M. van Koetsveld Ing et al., “Type I interferons in the treatment of pancreatic cancer: mechanisms of action and role of related Receptors,” Annals of Surgery, vol. 246, no. 2, pp. 259–268, 2007.
[3]  M. Marra, G. Salzano, C. Leonetti et al., “New self-assembly nanoparticles and stealth liposomes for the delivery of zoledronic acid: a comparative study,” Biotechnology Advances, vol. 30, no. 1, pp. 302–309, 2011.
[4]  M. Caraglia, G. Vitale, M. Marra, A. Budillon, P. Tagliaferri, and A. Abbruzzese, “Alpha-interferon and its effects on signalling pathways within cells,” Current Protein and Peptide Science, vol. 5, no. 6, pp. 475–485, 2004.
[5]  M. Marra, A. Lombardi, E. Agostinelli et al., “Bovine serum amine oxidase and spm potentiate docetaxel and interferon-α effects in inducing apoptosis on human cancer cells through the generation of oxidative stress,” Biochimica et Biophysica Acta, vol. 1783, no. 12, pp. 2269–2278, 2008.
[6]  M. Caraglia, M. Marra, P. Tagliaferri et al., “Emerging strategies to strengthen the anti-tumour activity of type I interferons: overcoming survival pathways,” Current Cancer Drug Targets, vol. 9, no. 5, pp. 690–704, 2009.
[7]  C. Jolly and R. I. Morimoto, “Role of the heat shock response and molecular chaperones in oncogenesis and cell death,” Journal of the National Cancer Institute, vol. 92, no. 19, pp. 1564–1572, 2000.
[8]  S. K. Calderwood, M. A. Khaleque, D. B. Sawyer, and D. R. Ciocca, “Heat shock proteins in cancer: chaperones of tumorigenesis,” Trends in Biochemical Sciences, vol. 31, no. 3, pp. 164–172, 2006.
[9]  C. Garrido, A. Fromentin, B. Bonnotte et al., “Heat shock protein 27 enhances the tumorigenicity of immunogenic rat colon carcinoma cell clones,” Cancer Research, vol. 58, no. 23, pp. 5495–5499, 1998.
[10]  S. Gurbuxani, J. M. Bruey, A. Fromentin et al., “Selective depletion of inducible HSP70 enhances immunogenicity of rat colon cancer cells,” Oncogene, vol. 20, no. 51, pp. 7478–7485, 2001.
[11]  C. De Simone, P. Ferranti, G. Picariello et al., “Peptides from water buffalo cheese whey induced senescence cell death via ceramide secretion in human colon adenocarcinoma cell line,” Molecular Nutrition and Food Research, vol. 55, no. 2, pp. 229–238, 2011.
[12]  R. Hayashi, Y. Ishii, H. Ochiai et al., “Suppression of heat shock protein 27 expression promotes 5-fluorouracil sensitivity in colon cancer cells in a xenograft model,” Oncology Reports, vol. 28, no. 4, pp. 1269–1274, 2012.
[13]  P. Mehlen, A. Mehlen, J. Godet, and A. P. Arrigo, “hsp27 as a switch between differentiation and apoptosis in murine embryonic stem cells,” Journal of Biological Chemistry, vol. 272, no. 50, pp. 31657–31665, 1997.
[14]  D. R. Ciocca, S. Oesterreich, G. C. Chamness, W. L. McGuire, and S. A. W. Fuqua, “Biological and clinical implications of heat shock protein 27 000 (Hsp27): a review,” Journal of the National Cancer Institute, vol. 85, no. 19, pp. 1558–1570, 1993.
[15]  P. Stiuso, G. Giuberti, A. Lombardi et al., “γ-Glutamyl 16-diaminopropane derivative of vasoactive intestinal peptide: a potent anti-oxidative agent for human epidermoid cancer cells,” Amino Acids, vol. 39, no. 3, pp. 661–670, 2010.
[16]  L. Vigh, B. Maresca, and J. L. Harwood, “Does the membrane's physical state control the expression of heat shock and other genes?” Trends in Biochemical Sciences, vol. 23, no. 10, pp. 369–374, 1998.
[17]  Z. T?r?k, N. M. Tsvetkova, G. Balogh et al., “Heat shock protein coinducers with no effect on protein denaturation specifically modulate the membrane lipid phase,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 6, pp. 3131–3136, 2003.
[18]  E. Nagy, Z. Balogi, I. Gombos et al., “Hyperfluidization-coupled membrane microdomain reorganization is linked to activation of the heat shock response in a murine melanoma cell line,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 19, pp. 7945–7950, 2007.
[19]  A. Budillon, F. Bruzzese, E. Di Gennaro, and M. Caraglia, “Multiple-target drugs: Inhibitors of heat shock protein 90 and of histone deacetylase,” Current Drug Targets, vol. 6, no. 3, pp. 337–351, 2005.
[20]  G. Misso, G. Giuberti, A. Lombardi et al., “Pharmacological inhibition of HSP90 and ras activity as a new strategy in the treatment of HNSCC,” Journal of Cellular Physiology, vol. 228, no. 1, pp. 130–141, 2013.
[21]  M. Caraglia, M. Marra, A. Budillon et al., “Chemotherapy regimen GOLF induces apoptosis in colon cancer cells through multi-chaperone complex inactivation and increased Raf-1 ubiquitin-dependent degradation,” Cancer Biology and Therapy, vol. 4, no. 10, pp. 1159–1167, 2005.
[22]  G. Vitale, S. Zappavigna, M. Marra et al., “The PPAR-gamma agonist troglitazone antagonizes survival pathways induced by STAT-3 in recombinant interferon-gamma treated pancreatic cancer cells,” Biotechnology Advances, vol. 30, no. 1, pp. 169–184, 2012.
[23]  G. L. Johnson and R. Lapadat, “Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases,” Science, vol. 298, no. 5600, pp. 1911–1912, 2002.
[24]  M. G. Wilkinson and J. B. A. Millar, “Control of the eukaryotic cell cycle by MAP kinase signaling pathways,” The FASEB Journal, vol. 14, no. 14, pp. 2147–2157, 2000.
[25]  S.-F. Chen, S. Nieh, S.-W. Jao et al., “Quercetin suppresses drug-resistant spheres via the p38 MAPK-Hsp27 apoptotic pathway in oral cancer cells,” PLoS ONE, vol. 7, no. 11, Article ID e49275, 2012.
[26]  I. Gomez-Monterrey, P. Campiglia, A. Bertamino et al., “A novel quinone-based derivative (DTNQ-Pro) induces apoptotic death via modulation/reduction of hsp expression in a human colon adenocarcinoma cell line,” British Journal of Pharmacology, vol. 160, no. 4, pp. 931–940, 2010.
[27]  C. De Simone, G. Picariello, G. Mamone et al., “Characterisation and cytomodulatory properties of peptides from Mozzarella di Bufala Campana cheese whey,” Journal of Peptide Science, vol. 15, no. 3, pp. 251–258, 2009.
[28]  F. Herz, A. Schermer, M. Halwer, and L. H. Bogart, “Alkaline phosphatase in HT-29, a human colon cancer cell line: Influence of sodium butyrate and hyperosmolality,” Archives of Biochemistry and Biophysics, vol. 210, no. 2, pp. 581–591, 1981.
[29]  M. M. Bradford, “A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein dye binding,” Analytical Biochemistry, vol. 72, no. 1-2, pp. 248–254, 1976.
[30]  Q. M. Ding, T. C. Ko, and B. Mark Evers, “Caco-2 intestinal cell differentiation is associated with G1 arrest and suppression of CDK2 and CDK4,” American Journal of Physiology, vol. 275, no. 5, pp. C1193–C1200, 1998.
[31]  H. Matsumoto, R. H. Erickson, J. R. Gum, M. Yoshioka, E. Gum, and Y. S. Kim, “Biosynthesis of alkaline phosphatase during differentiation of the human colon cancer cell line Caco-2,” Gastroenterology, vol. 98, no. 5 I, pp. 1199–1207, 1990.
[32]  D. A. Wink and J. B. Mitchell, “Chemical biology of nitric oxide: Insights into regulatory, cytotoxic, and cytoprotective mechanisms of nitric oxide,” Free Radical Biology and Medicine, vol. 25, no. 4-5, pp. 434–456, 1998.
[33]  H. Huang and D. J. Tindall, “Dynamic FoxO transcription factors,” Journal of Cell Science, vol. 120, no. 15, pp. 2479–2487, 2007.
[34]  S. Popat, A. Matakidou, and R. S. Houlston, “Thymidylate-synthase expression and prognosis in colorectal cancer: a systematic review and meta-analysis,” Journal of Clinical Oncology, vol. 22, no. 3, pp. 529–536, 2004.
[35]  L. Whitesell and S. L. Lindquist, “HSP90 and the chaperoning of cancer,” Nature Reviews Cancer, vol. 5, no. 10, pp. 761–772, 2005.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413