Skip to main content
Top
Published in: Cellular Oncology 3/2014

01-06-2014 | Review

Targeting epigenetic mechanisms and microRNAs by aspirin and other non steroidal anti-inflammatory agents - implications for cancer treatment and chemoprevention

Author: Eugenia Yiannakopoulou

Published in: Cellular Oncology | Issue 3/2014

Login to get access

Abstract

Background

Epigenetic processes and miRNAs have been recognized as new targets for anticancer drug design. However, old multi-target drugs such as aspirin may also target epigenetic processes.

Aim

This review aims to provide an overview of our current knowledge on the modulation of epigenetic processes by aspirin and other non steroidal anti-inflammatory agents (NSAIDs) and their implications for cancer treatment and chemoprevention.

Synthesis

In vitro and in vivo studies, as well as primary patient data, suggest that aspirin and other NSAIDs reverse tumour suppressor gene hypermethylation in cancer tissues. It must be emphasized that, at this point in time, patient data are limited and DNA hypermethylation reversal has been investigated, but not tumour suppressor gene activation. In addition, evidence from experimental and patient data suggests that aspirin and NSAIDs may also reverse global DNA hypomethylation. At the histone level, both induction and inhibition of deacetylases by aspirin have been reported. Also, direct acetylation of histones by aspirin has been reported, while the natural salicylate anacardic acid has been found to inhibit histone acetyltransferase p300 both in vitro and in vivo, and to regulate gene expression through modulation of histone acetylation. Salicylates and other NSAIDs may also down-regulate miRNAs with oncogene-like functions or up-regulate miRNAs with tumour suppressor-like functions. Up till now, clinical trials have been aimed at investigating the effect of salicylates and NSAIDs on a limited number of miRNAs.

Conclusion

So, although the existing evidence is still limited, evidence is accumulating that epigenetic targets may represent nodal targets for the anti-proliferative actions of salicylates and NSAIDs. This, in turn, may have implications for cancer chemoprevention and treatment. Undoubtedly, this notion requires further investigation, but if proved correct, it could lead to the design of less toxic agents that target epigenetic processes as part of existing or novel multi-targeted treatment modalities.
Literature
1.
go back to reference D. Nebert, G. Zhang, E. Vesell, From human genetics and genomics to pharmacogenetics and pharmacogenomics: past lessons, future directions. Drug Metab Rev 40, 187–224 (2008)PubMedCentralPubMedCrossRef D. Nebert, G. Zhang, E. Vesell, From human genetics and genomics to pharmacogenetics and pharmacogenomics: past lessons, future directions. Drug Metab Rev 40, 187–224 (2008)PubMedCentralPubMedCrossRef
3.
go back to reference J.G. Martínez, J. Pérez-Escuredo, P. Castro-Santos, C.A. Marcos, J.L. Pendás, M.F. Fraga, Hermsen. M.A. Hypomethylation of LINE-1, and not centromeric SAT-α, is associated with centromeric instability in head and neck squamous cell carcinoma. Cell Oncol 35, 259–67 (2012)CrossRef J.G. Martínez, J. Pérez-Escuredo, P. Castro-Santos, C.A. Marcos, J.L. Pendás, M.F. Fraga, Hermsen. M.A. Hypomethylation of LINE-1, and not centromeric SAT-α, is associated with centromeric instability in head and neck squamous cell carcinoma. Cell Oncol 35, 259–67 (2012)CrossRef
4.
go back to reference S. Babashah, M. Sadeghizadeh, M.R. Tavirani, S. Farivar, Soleimani. M. Aberrant microRNA expression and its implications in the pathogenesis of leukemias. Cell Oncol 35, 317–34 (2012)CrossRef S. Babashah, M. Sadeghizadeh, M.R. Tavirani, S. Farivar, Soleimani. M. Aberrant microRNA expression and its implications in the pathogenesis of leukemias. Cell Oncol 35, 317–34 (2012)CrossRef
5.
go back to reference R. Nagadia, P. Pandit, W.B. Coman, J. Cooper-White, C. Punyadeera, miRNAs in head and neck cancer revisited. Cell Oncol 36, 1–7 (2013)CrossRef R. Nagadia, P. Pandit, W.B. Coman, J. Cooper-White, C. Punyadeera, miRNAs in head and neck cancer revisited. Cell Oncol 36, 1–7 (2013)CrossRef
6.
go back to reference C. Lema, M. Cunningham, MicroRNAs and their implications in toxicological research. Toxicol Lett 198, 100–105 (2010)PubMedCrossRef C. Lema, M. Cunningham, MicroRNAs and their implications in toxicological research. Toxicol Lett 198, 100–105 (2010)PubMedCrossRef
7.
go back to reference P.J. Mishra, P.J. Mishra, D. Barenjee, J.R. Bertino, MiRNSPs or MiR polymorphisms, new players in microRNA mediated regulation of the cell. Cell Cycle 7, 853–858 (2008)PubMedCrossRef P.J. Mishra, P.J. Mishra, D. Barenjee, J.R. Bertino, MiRNSPs or MiR polymorphisms, new players in microRNA mediated regulation of the cell. Cell Cycle 7, 853–858 (2008)PubMedCrossRef
8.
go back to reference P.J. Mishra, J.R. Bertino, MicroRNA polymorphisms: the future of pharmacogenomics, molecular epidemiology and personalized medicine. Pharmacogenomics 10, 399–416 (2009)PubMedCentralPubMedCrossRef P.J. Mishra, J.R. Bertino, MicroRNA polymorphisms: the future of pharmacogenomics, molecular epidemiology and personalized medicine. Pharmacogenomics 10, 399–416 (2009)PubMedCentralPubMedCrossRef
9.
go back to reference E.D. Wiklund, J. Kjems, S.J. Clark, Epigenetic architecture and miRNA: reciprocal regulators. Epigenomics 6, 823–840 (2010)CrossRef E.D. Wiklund, J. Kjems, S.J. Clark, Epigenetic architecture and miRNA: reciprocal regulators. Epigenomics 6, 823–840 (2010)CrossRef
10.
go back to reference F.A. Duijkers, R.X. de Menezes, I.J. Goossens-Beumer, D.J. Stumpel, P. Admiraal, R. Pieters, J.P. Meijerink, M.M. van Noesel, Epigenetic drug combination induces genome-wide demethylation and altered gene expression in neuro-ectodermal tumor-derived cell lines. Cell Oncol 36, 351–62 (2013)CrossRef F.A. Duijkers, R.X. de Menezes, I.J. Goossens-Beumer, D.J. Stumpel, P. Admiraal, R. Pieters, J.P. Meijerink, M.M. van Noesel, Epigenetic drug combination induces genome-wide demethylation and altered gene expression in neuro-ectodermal tumor-derived cell lines. Cell Oncol 36, 351–62 (2013)CrossRef
11.
go back to reference J.P. Issa, H.M. Kantarjian, Targeting DNA methylation Clin Cancer Res 15, 3938–46 (2009)CrossRef J.P. Issa, H.M. Kantarjian, Targeting DNA methylation Clin Cancer Res 15, 3938–46 (2009)CrossRef
12.
go back to reference J. García-Quiroz, J. Camacho, Astemizole: an old anti-histamine as a new promising anti-cancer drug. Anticancer Agents Med Chem 11, 307–14 (2011)PubMedCrossRef J. García-Quiroz, J. Camacho, Astemizole: an old anti-histamine as a new promising anti-cancer drug. Anticancer Agents Med Chem 11, 307–14 (2011)PubMedCrossRef
13.
go back to reference C. Zhao, X. Bu, Promoter methylation of tumor-related genes in gastric carcinogenesis. Histol Histopathol 27, 1271–82 (2012)PubMed C. Zhao, X. Bu, Promoter methylation of tumor-related genes in gastric carcinogenesis. Histol Histopathol 27, 1271–82 (2012)PubMed
14.
go back to reference T.M. Brasky, M.R. Bonner, K.B. Moysich, C.B. Ambrosone, J. Nie, M.H. Tao, S.B. Edge, B.V. Kallakury, H.M. Ochs-Balcom, C. Marian, M. Trevisan, P.G. Shields, J.L. Freudenheim, Non steroidal anti-inflammatory drug use (NSAID) and breast cancer risk in the western New York exposures and breast cancer (WEB) study. Cancer Causes Control 9, 1503–1512 (2010)CrossRef T.M. Brasky, M.R. Bonner, K.B. Moysich, C.B. Ambrosone, J. Nie, M.H. Tao, S.B. Edge, B.V. Kallakury, H.M. Ochs-Balcom, C. Marian, M. Trevisan, P.G. Shields, J.L. Freudenheim, Non steroidal anti-inflammatory drug use (NSAID) and breast cancer risk in the western New York exposures and breast cancer (WEB) study. Cancer Causes Control 9, 1503–1512 (2010)CrossRef
15.
go back to reference Li, X.; Gao, L.; Cui, Q.; Gary, B.D.; Dyess, D.L.; Taylor, W.; Shevde, L.A.; Samant, R.S.; Dean-Colomb, W.; Piazza, G.A.; Xi, Y. Sulindac inhibits tumor cell invasion by suppressing NF-κB-mediated transcription of microRNAs. Oncogene 31, 4979–4986 (2012) Li, X.; Gao, L.; Cui, Q.; Gary, B.D.; Dyess, D.L.; Taylor, W.; Shevde, L.A.; Samant, R.S.; Dean-Colomb, W.; Piazza, G.A.; Xi, Y. Sulindac inhibits tumor cell invasion by suppressing NF-κB-mediated transcription of microRNAs. Oncogene 31, 4979–4986 (2012)
16.
go back to reference E. Yiannakopoulou, Modulation of lymphangiogenesis: a New target for aspirin and other Nonsteroidal anti-inflammatory agents? a systematic review. J Clin Pharmacol 52, 1749–54 (2011)PubMedCrossRef E. Yiannakopoulou, Modulation of lymphangiogenesis: a New target for aspirin and other Nonsteroidal anti-inflammatory agents? a systematic review. J Clin Pharmacol 52, 1749–54 (2011)PubMedCrossRef
17.
go back to reference M. Dovizio, A. Bruno, S. Tacconelli, P. Patrignani, Mode of action of aspirin as a chemopreventive agent. Recent Results Cancer Res 191, 39–65 (2013)PubMedCrossRef M. Dovizio, A. Bruno, S. Tacconelli, P. Patrignani, Mode of action of aspirin as a chemopreventive agent. Recent Results Cancer Res 191, 39–65 (2013)PubMedCrossRef
18.
go back to reference J. Worm, P. Guldberg, DNA methylation: an epigenetic pathway to cancer and a promising target for anticancer therapy. J Oral Pathol Med 31, 443–9 (2002)PubMedCrossRef J. Worm, P. Guldberg, DNA methylation: an epigenetic pathway to cancer and a promising target for anticancer therapy. J Oral Pathol Med 31, 443–9 (2002)PubMedCrossRef
19.
go back to reference J. Paluszczak, V. Krajka-Kuźniak, W. Baer-Dubowska, The effect of dietary polyphenols on the epigenetic regulation of gene expression in MCF7 breast cancer cells. Toxicol Lett 192, 119–125 (2010)PubMedCrossRef J. Paluszczak, V. Krajka-Kuźniak, W. Baer-Dubowska, The effect of dietary polyphenols on the epigenetic regulation of gene expression in MCF7 breast cancer cells. Toxicol Lett 192, 119–125 (2010)PubMedCrossRef
20.
go back to reference J.L. Medina-Franco, T. Caulfield, Advances in the computational development of DNA methyltransferase inhibitors. Drug Discov Today 16, 418–425 (2011)PubMedCrossRef J.L. Medina-Franco, T. Caulfield, Advances in the computational development of DNA methyltransferase inhibitors. Drug Discov Today 16, 418–425 (2011)PubMedCrossRef
21.
go back to reference D. Kuck, N. Singh, F. Lyko, J.L. Medina-Franco, Novel and selective DNA methyltransferase inhibitors: Docking-based virtual screening and experimental evaluation. Bioorg Med Chem 18, 822–829 (2010)PubMedCrossRef D. Kuck, N. Singh, F. Lyko, J.L. Medina-Franco, Novel and selective DNA methyltransferase inhibitors: Docking-based virtual screening and experimental evaluation. Bioorg Med Chem 18, 822–829 (2010)PubMedCrossRef
22.
go back to reference J. Yoo, J.L. Medina Franco, Trimethylaurintricarboxylic acid inhibits human DNA methyltransferase 1: insights from enzymatic and molecular modeling studies. J Mol Model 18, 1583–1589 (2012)PubMedCrossRef J. Yoo, J.L. Medina Franco, Trimethylaurintricarboxylic acid inhibits human DNA methyltransferase 1: insights from enzymatic and molecular modeling studies. J Mol Model 18, 1583–1589 (2012)PubMedCrossRef
23.
go back to reference M.R. Pan, H.C. Chang, L.Y. Chuang, W.C. Hung, The nonsteroidal anti-inflammatory drug NS398 reactivates SPARC expression via promoter demethylation to attenuate invasiveness of lung cancer cells. Exp Biol Med (Maywood) 233, 456–462 (2008)CrossRef M.R. Pan, H.C. Chang, L.Y. Chuang, W.C. Hung, The nonsteroidal anti-inflammatory drug NS398 reactivates SPARC expression via promoter demethylation to attenuate invasiveness of lung cancer cells. Exp Biol Med (Maywood) 233, 456–462 (2008)CrossRef
24.
go back to reference T. Tahara, T. Shibata, M. Nakamura, H. Yamashita, D. Yoshioka, M. Okubo, N. Maruyama, T. Kamano, Y. Kamiya, H. Fujita, M. Nagasaka, M. Iwata, K. Takahama, M. Watanabe, I. Hirata, T. Arisawa, Chronic aspirin use suppresses CDH1 methylation in human gastric mucosa. Dig Dis Sci 55, 54–9 (2010)PubMedCrossRef T. Tahara, T. Shibata, M. Nakamura, H. Yamashita, D. Yoshioka, M. Okubo, N. Maruyama, T. Kamano, Y. Kamiya, H. Fujita, M. Nagasaka, M. Iwata, K. Takahama, M. Watanabe, I. Hirata, T. Arisawa, Chronic aspirin use suppresses CDH1 methylation in human gastric mucosa. Dig Dis Sci 55, 54–9 (2010)PubMedCrossRef
25.
go back to reference T. Tahara, T. Shibata, H. Yamashita, M. Nakamura, D. Yoshioka, M. Okubo, I. Hirata, T. Arisawa, Chronic nonsteroidal anti-inflammatory drug (NSAID) use suppresses multiple CpG islands hyper methylation (CIHM) of tumor suppressor genes in the human gastric mucosa. Cancer Sci 100, 1192–1197 (2009)PubMedCrossRef T. Tahara, T. Shibata, H. Yamashita, M. Nakamura, D. Yoshioka, M. Okubo, I. Hirata, T. Arisawa, Chronic nonsteroidal anti-inflammatory drug (NSAID) use suppresses multiple CpG islands hyper methylation (CIHM) of tumor suppressor genes in the human gastric mucosa. Cancer Sci 100, 1192–1197 (2009)PubMedCrossRef
26.
go back to reference M.L. Slattery, R.K. Wolff, J. Herrick, B.J. Caan, W. Samowitz, Tumor markers and rectal cancer: support for an inflammation-related pathway. Int J Cancer 125, 1698–1704 (2009)PubMedCentralPubMedCrossRef M.L. Slattery, R.K. Wolff, J. Herrick, B.J. Caan, W. Samowitz, Tumor markers and rectal cancer: support for an inflammation-related pathway. Int J Cancer 125, 1698–1704 (2009)PubMedCentralPubMedCrossRef
27.
go back to reference M.L. Slattery, K. Curtin, C. Sweeney, T.R. Levin, J. Potter, R.K. Wolff, H. Albertsen, W.S. Samowitz, Diet and lifestyle factor associations with CpG island methylator phenotype and BRAF mutations in colon cancer. Int J Cancer 120, 656–663 (2007)PubMedCrossRef M.L. Slattery, K. Curtin, C. Sweeney, T.R. Levin, J. Potter, R.K. Wolff, H. Albertsen, W.S. Samowitz, Diet and lifestyle factor associations with CpG island methylator phenotype and BRAF mutations in colon cancer. Int J Cancer 120, 656–663 (2007)PubMedCrossRef
28.
go back to reference E.I. Heath, M.I. Canto, S. Piantadosi, E. Montgomery, W.M. Weinstein, J.G. Herman, A.J. Dannenberg, V.W. Yang, A.O. Shar, E. Hawk, A.A. Forastiere, Chemoprevention for Barrett’s esophagus trial research group. Secondary chemoprevention of Barrett’s esophagus with celecoxib: results of a randomized trial. J. Natl. Cancer Risk 99, 545–557 (2007) E.I. Heath, M.I. Canto, S. Piantadosi, E. Montgomery, W.M. Weinstein, J.G. Herman, A.J. Dannenberg, V.W. Yang, A.O. Shar, E. Hawk, A.A. Forastiere, Chemoprevention for Barrett’s esophagus trial research group. Secondary chemoprevention of Barrett’s esophagus with celecoxib: results of a randomized trial. J. Natl. Cancer Risk 99, 545–557 (2007)
29.
go back to reference K. Wallace, M.V. Grau, A.J. Levine, L. Shen, R. Hamdan, X. Chen, J. Gui, R.W. Haile, E.L. Barry, D. Ahnen, G. McKeown-Eyssen, J.A. Baron, J.P. Issa, Association between folate levels and CpG Island hypermethylation in normal colorectal mucosa. Cancer Prev Res (Phila) 3, 1552–1564 (2010)CrossRef K. Wallace, M.V. Grau, A.J. Levine, L. Shen, R. Hamdan, X. Chen, J. Gui, R.W. Haile, E.L. Barry, D. Ahnen, G. McKeown-Eyssen, J.A. Baron, J.P. Issa, Association between folate levels and CpG Island hypermethylation in normal colorectal mucosa. Cancer Prev Res (Phila) 3, 1552–1564 (2010)CrossRef
30.
go back to reference J.C. Figueiredo, M.V. Grau, K. Wallace, A.J. Levine, L. Shen, R. Hamdan, X. Chen, R.S. Bresalier, G. McKeown-Eyssen, R.W. Haile, J.A. Baron, J.P. Issa, Global DNA hypomethylation (LINE-1) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer Epidemiol Biomarkers Prev 18, 1041–1049 (2009)PubMedCentralPubMedCrossRef J.C. Figueiredo, M.V. Grau, K. Wallace, A.J. Levine, L. Shen, R. Hamdan, X. Chen, R.S. Bresalier, G. McKeown-Eyssen, R.W. Haile, J.A. Baron, J.P. Issa, Global DNA hypomethylation (LINE-1) in the normal colon and lifestyle characteristics and dietary and genetic factors. Cancer Epidemiol Biomarkers Prev 18, 1041–1049 (2009)PubMedCentralPubMedCrossRef
31.
go back to reference L. Tao, W. Wang, P.M. Kramer, R.A. Lubet, V.E. Steele, M.A. Pereira, Modulation of DNA hypomethylation as a surrogate endpoint biomarker for chemoprevention, of colon cancer. Mol Carcinogen 39, 79–842 (2004)CrossRef L. Tao, W. Wang, P.M. Kramer, R.A. Lubet, V.E. Steele, M.A. Pereira, Modulation of DNA hypomethylation as a surrogate endpoint biomarker for chemoprevention, of colon cancer. Mol Carcinogen 39, 79–842 (2004)CrossRef
32.
go back to reference M.A. Pereira, L. Tao, W. Wang, Y. Li, A. Umar, V.E. Steele, Modulation by celecoxib and difluoromethylornithine of the methylation of DNA and the estrogen receptor-alpha gene in rat colon tumors. Carcinogenesis 25, 1917–1923 (2004)PubMedCrossRef M.A. Pereira, L. Tao, W. Wang, Y. Li, A. Umar, V.E. Steele, Modulation by celecoxib and difluoromethylornithine of the methylation of DNA and the estrogen receptor-alpha gene in rat colon tumors. Carcinogenesis 25, 1917–1923 (2004)PubMedCrossRef
33.
go back to reference R. Shen, L. Tao, Y. Xu, S. Chang, J. Van Brocklyn, J.X. Gao, Reversibility of aberrant global DNA and estrogen receptor-alpha gene methylation distinguishes colorectal precancer from cancer. Int J Clin Exp Pathol 2, 21–23 (2009)PubMedCentralPubMed R. Shen, L. Tao, Y. Xu, S. Chang, J. Van Brocklyn, J.X. Gao, Reversibility of aberrant global DNA and estrogen receptor-alpha gene methylation distinguishes colorectal precancer from cancer. Int J Clin Exp Pathol 2, 21–23 (2009)PubMedCentralPubMed
34.
go back to reference R.M. Eglen, T. Reisine, Screening for compounds that modulate epigenetic regulation of the transcriptome: an overview. J Biomol Screen 16, 1137–1152 (2011)PubMedCrossRef R.M. Eglen, T. Reisine, Screening for compounds that modulate epigenetic regulation of the transcriptome: an overview. J Biomol Screen 16, 1137–1152 (2011)PubMedCrossRef
35.
go back to reference A. Mai, L. Altucci, Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 41, 199–213 (2009)PubMedCrossRef A. Mai, L. Altucci, Epi-drugs to fight cancer: from chemistry to cancer treatment, the road ahead. Int J Biochem Cell Biol 41, 199–213 (2009)PubMedCrossRef
36.
go back to reference P. Kamble, K. Selvarajan, C. Aluganti Narasimhulu, M. Nandave, S. Parthasarathy, Aspirin may promote mitochondrial biogenesis via the production of hydrogen peroxide and the induction of Sirtuin1/PGC-1α genes. Eur J Pharmacol 699, 55–61 (2013)PubMedCentralPubMedCrossRef P. Kamble, K. Selvarajan, C. Aluganti Narasimhulu, M. Nandave, S. Parthasarathy, Aspirin may promote mitochondrial biogenesis via the production of hydrogen peroxide and the induction of Sirtuin1/PGC-1α genes. Eur J Pharmacol 699, 55–61 (2013)PubMedCentralPubMedCrossRef
37.
go back to reference F. Di Renzo, G. Cappelletti, M.L. Broccia, E. Giavini, E. Menegola, The inhibition of embryonic histone deacetylases as the possible mechanism accounting for axial skeletal malformations induced by sodium salicylate. Toxicol Sci 104, 397–404 (2008)PubMedCrossRef F. Di Renzo, G. Cappelletti, M.L. Broccia, E. Giavini, E. Menegola, The inhibition of embryonic histone deacetylases as the possible mechanism accounting for axial skeletal malformations induced by sodium salicylate. Toxicol Sci 104, 397–404 (2008)PubMedCrossRef
38.
go back to reference J. Sonnemann, I. Hüls, M. Sigler, C.D. Palani, T.T. le Hong, U. Völker, H.K. Kroemer, J.F. Beck, Histone deacetylase inhibitors and aspirin interact synergistically to induce cell death in ovarian cancer cells. Oncol Rep 20, 219–224 (2008)PubMed J. Sonnemann, I. Hüls, M. Sigler, C.D. Palani, T.T. le Hong, U. Völker, H.K. Kroemer, J.F. Beck, Histone deacetylase inhibitors and aspirin interact synergistically to induce cell death in ovarian cancer cells. Oncol Rep 20, 219–224 (2008)PubMed
39.
go back to reference K. Deckmann, F. Rörsch, G. Geisslinger, S. Grösch, Dimethylcelecoxib induces an inhibitory complex consisting of HDAC1/NF-κB(p65)RelA leading to transcriptional downregulation of mPGES-1 and EGR1. Cell Signal 24, 460–467 (2012)PubMedCrossRef K. Deckmann, F. Rörsch, G. Geisslinger, S. Grösch, Dimethylcelecoxib induces an inhibitory complex consisting of HDAC1/NF-κB(p65)RelA leading to transcriptional downregulation of mPGES-1 and EGR1. Cell Signal 24, 460–467 (2012)PubMedCrossRef
40.
go back to reference H. Yoshioka, T. Watanabe, T.E. Eling, Nonsteroidal anti-inflammatory drug-activated gene (NAG-1/GDF15) expression is increased by the histone deacetylase inhibitor trichostatin A. J Biol Chem 283, 33129–33137 (2008)PubMedCentralPubMedCrossRef H. Yoshioka, T. Watanabe, T.E. Eling, Nonsteroidal anti-inflammatory drug-activated gene (NAG-1/GDF15) expression is increased by the histone deacetylase inhibitor trichostatin A. J Biol Chem 283, 33129–33137 (2008)PubMedCentralPubMedCrossRef
41.
go back to reference S. Marimuthu, R.S. Chivukula, L.F. Alfonso, M. Moridani, F.K. Hagen, G.J. Bhat, Aspirin acetylates multiple cellular proteins in HCT-116 colon cancer cells: Identification of novel targets. Int J Oncol 39, 1273–1283 (2011)PubMed S. Marimuthu, R.S. Chivukula, L.F. Alfonso, M. Moridani, F.K. Hagen, G.J. Bhat, Aspirin acetylates multiple cellular proteins in HCT-116 colon cancer cells: Identification of novel targets. Int J Oncol 39, 1273–1283 (2011)PubMed
42.
go back to reference S.B. Jung, C.S. Kim, A. Naqvi, T. Yamamori, I. Mattagajasingh, T.A. Hoffman, M.P. Cole, A. Kumar, J.S. Dericco, B.H. Jeon, K. Irani, Histone deacetylase 3 antagonizes aspirin-stimulated endothelial nitric oxide production by reversing aspirin-induced lysine acetylation of endothelial nitric oxide synthase. Circ Res 107, 877–87 (2010)PubMedCentralPubMedCrossRef S.B. Jung, C.S. Kim, A. Naqvi, T. Yamamori, I. Mattagajasingh, T.A. Hoffman, M.P. Cole, A. Kumar, J.S. Dericco, B.H. Jeon, K. Irani, Histone deacetylase 3 antagonizes aspirin-stimulated endothelial nitric oxide production by reversing aspirin-induced lysine acetylation of endothelial nitric oxide synthase. Circ Res 107, 877–87 (2010)PubMedCentralPubMedCrossRef
43.
go back to reference J. Tan, B. Chen, L. He, Y. Tang, Z. Jiang, G. Yin, J. Wang, X. Jiang, Anacardic acid (6-pentadecylsalicylic acid) induces apoptosis of prostate cancer cells through inhibition of androgen receptor and activation of p53 signaling. Chin J Cancer Res 24, 275–83 (2012)PubMedCentralPubMedCrossRef J. Tan, B. Chen, L. He, Y. Tang, Z. Jiang, G. Yin, J. Wang, X. Jiang, Anacardic acid (6-pentadecylsalicylic acid) induces apoptosis of prostate cancer cells through inhibition of androgen receptor and activation of p53 signaling. Chin J Cancer Res 24, 275–83 (2012)PubMedCentralPubMedCrossRef
44.
go back to reference B. Sung, M.K. Pandey, K.S. Ahn, T. Yi, M.M. Chaturvedi, M. Liu, B.B. Aggarwal, Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-kappaB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-kappaBalpha kinase, leading to potentiation of apoptosis. Blood 111, 4880–4891 (2008)PubMedCentralPubMedCrossRef B. Sung, M.K. Pandey, K.S. Ahn, T. Yi, M.M. Chaturvedi, M. Liu, B.B. Aggarwal, Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-kappaB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-kappaBalpha kinase, leading to potentiation of apoptosis. Blood 111, 4880–4891 (2008)PubMedCentralPubMedCrossRef
45.
go back to reference Y. Sun, X. Jiang, S. Chen, B.D. Price, Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation. FEBS Lett 580, 4353–4356 (2006)PubMedCrossRef Y. Sun, X. Jiang, S. Chen, B.D. Price, Inhibition of histone acetyltransferase activity by anacardic acid sensitizes tumor cells to ionizing radiation. FEBS Lett 580, 4353–4356 (2006)PubMedCrossRef
46.
go back to reference Y. Kim, K. Kim, D. Park, E. Lee, H. Lee, Y.S. Lee, J. Choe, Y.M. Kim, D. Jeoung, DNA methyl transferase I acts as a negative regulator of allergic skin inflammation. Mol Immunol 53, 1–14 (2013)PubMedCrossRef Y. Kim, K. Kim, D. Park, E. Lee, H. Lee, Y.S. Lee, J. Choe, Y.M. Kim, D. Jeoung, DNA methyl transferase I acts as a negative regulator of allergic skin inflammation. Mol Immunol 53, 1–14 (2013)PubMedCrossRef
47.
go back to reference A. Koornneef, K. Rindermann, C. Gatz, C.M. Pieterse, Histone modifications do not play a major role in salicylate-mediated suppression of jasmonate-induced PDF1.2 gene expression. Commun Integr Biol 1, 143–5 (2008)PubMedCentralPubMedCrossRef A. Koornneef, K. Rindermann, C. Gatz, C.M. Pieterse, Histone modifications do not play a major role in salicylate-mediated suppression of jasmonate-induced PDF1.2 gene expression. Commun Integr Biol 1, 143–5 (2008)PubMedCentralPubMedCrossRef
48.
go back to reference T. Tanaka, A. Kurose, H.D. Halicka, X. Huang, F. Traganos, Z. Darzynkiewicz, Nitrogen oxide-releasing aspirin induces histone H2AX phosphorylation, ATM activation and apoptosis preferentially in S-phase cells: involvement of reactive oxygen species. Cell Cycle 5, 1669–74 (2006)PubMedCrossRef T. Tanaka, A. Kurose, H.D. Halicka, X. Huang, F. Traganos, Z. Darzynkiewicz, Nitrogen oxide-releasing aspirin induces histone H2AX phosphorylation, ATM activation and apoptosis preferentially in S-phase cells: involvement of reactive oxygen species. Cell Cycle 5, 1669–74 (2006)PubMedCrossRef
49.
go back to reference M. Yanokura, K. Banno, Y. Kobayashi, I. Kisu, A. Ueki, A. Ono, K. Masuda, H. Nomura, A. Hirasawa, N. Susumu, D. Aoki, MicroRNA and endometrial cancer: Roles of small RNAs in human tumors and clinical applications (Review). Oncol Lett 1, 935–940 (2010)PubMedCentralPubMed M. Yanokura, K. Banno, Y. Kobayashi, I. Kisu, A. Ueki, A. Ono, K. Masuda, H. Nomura, A. Hirasawa, N. Susumu, D. Aoki, MicroRNA and endometrial cancer: Roles of small RNAs in human tumors and clinical applications (Review). Oncol Lett 1, 935–940 (2010)PubMedCentralPubMed
50.
go back to reference F. Lan, X. Yue, L. Han, Z. Shi, Y. Yang, P. Pu, Z. Yao, C. Kang, Genome-wide identification of TCF7L2/TCF4 target miRNAs reveals a role for miR-21 in Wnt-driven epithelial cancer. Int J Oncol 40, 519–526 (2012)PubMed F. Lan, X. Yue, L. Han, Z. Shi, Y. Yang, P. Pu, Z. Yao, C. Kang, Genome-wide identification of TCF7L2/TCF4 target miRNAs reveals a role for miR-21 in Wnt-driven epithelial cancer. Int J Oncol 40, 519–526 (2012)PubMed
51.
go back to reference Y. Chen, W. Liu, T. Chao, Y. Zhang, X. Yan, Y. Gong, B. Qiang, J. Yuan, M. Sun, X. Peng, MicroRNA-21 down-regulates the expression of tumor suppressor PDCD4 in human glioblastoma cell T98G. Cancer Lett 272, 197–205 (2008)PubMedCrossRef Y. Chen, W. Liu, T. Chao, Y. Zhang, X. Yan, Y. Gong, B. Qiang, J. Yuan, M. Sun, X. Peng, MicroRNA-21 down-regulates the expression of tumor suppressor PDCD4 in human glioblastoma cell T98G. Cancer Lett 272, 197–205 (2008)PubMedCrossRef
52.
go back to reference F. Meng, R. Henson, H. Wehbe-Janek, K. Ghoshal, S.T. Jacob, T. Patel, MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 133, 647–658 (2007)PubMedCrossRef F. Meng, R. Henson, H. Wehbe-Janek, K. Ghoshal, S.T. Jacob, T. Patel, MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 133, 647–658 (2007)PubMedCrossRef
53.
go back to reference Q. Yao, H. Xu, Q.Q. Zhang, H. Zhou, L.H. Qu, MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells. Biochem Biophys Res Commun 388, 539–542 (2009)PubMedCrossRef Q. Yao, H. Xu, Q.Q. Zhang, H. Zhou, L.H. Qu, MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells. Biochem Biophys Res Commun 388, 539–542 (2009)PubMedCrossRef
54.
go back to reference Sicard, F.; Gayral, M.; Lulka, H.; Buscail, L.; Cordelier, P. Targeting miR-21 for the Therapy of Pancreatic Cancer. Mol Ther, 2013, doi: 10.1038/mt.2013.35 Sicard, F.; Gayral, M.; Lulka, H.; Buscail, L.; Cordelier, P. Targeting miR-21 for the Therapy of Pancreatic Cancer. Mol Ther, 2013, doi: 10.1038/mt.2013.35
55.
go back to reference Y. Saito, H. Suzuki, H. Imaeda, J. Matsuzaki, K. Hirata, H. Tsugawa, S. Hibino, Y. Kanai, H. Saito, T. Hibi, The tumor suppressor microRNA-29c is downregulated and restored by celecoxib in human gastric cancer cells. Int J Cancer 132, 1751–1760 (2013)PubMedCrossRef Y. Saito, H. Suzuki, H. Imaeda, J. Matsuzaki, K. Hirata, H. Tsugawa, S. Hibino, Y. Kanai, H. Saito, T. Hibi, The tumor suppressor microRNA-29c is downregulated and restored by celecoxib in human gastric cancer cells. Int J Cancer 132, 1751–1760 (2013)PubMedCrossRef
56.
go back to reference W.C. Chen, M.S. Lin, Y.L. Ye, H.J. Gao, Z.Y. Song, X.Y. Shen, microRNA expression pattern and its alteration following celecoxib intervention in human colorectal cancer. Exp Ther Med 3, 1039–1048 (2012)PubMedCentralPubMed W.C. Chen, M.S. Lin, Y.L. Ye, H.J. Gao, Z.Y. Song, X.Y. Shen, microRNA expression pattern and its alteration following celecoxib intervention in human colorectal cancer. Exp Ther Med 3, 1039–1048 (2012)PubMedCentralPubMed
57.
go back to reference J. Tan, L. Fan, J.J. Mao, B. Chen, L. Zheng, T. Zhang, T. Li, J. Duan, Y. Duan, Z. Jin, W. Kuang, Restoration of miR-34a in p53 deficient cells unexpectedly promotes the cell survival by increasing NFκB activity. J Cell Biochem 113, 2903–8 (2012)PubMedCrossRef J. Tan, L. Fan, J.J. Mao, B. Chen, L. Zheng, T. Zhang, T. Li, J. Duan, Y. Duan, Z. Jin, W. Kuang, Restoration of miR-34a in p53 deficient cells unexpectedly promotes the cell survival by increasing NFκB activity. J Cell Biochem 113, 2903–8 (2012)PubMedCrossRef
58.
go back to reference E.D. Eliseeva, V. Valkov, M. Jung, M.O. Jung, Characterization of novel inhibitors of histone acetyltransferases. Mol Cancer Ther 6, 2391–2398 (2007)PubMedCrossRef E.D. Eliseeva, V. Valkov, M. Jung, M.O. Jung, Characterization of novel inhibitors of histone acetyltransferases. Mol Cancer Ther 6, 2391–2398 (2007)PubMedCrossRef
59.
go back to reference A.R. Cyr, F.E. Domann, The redox basis of epigenetic modifications: from mechanisms to functional consequences. Antioxid Redox Signal 15, 551–589 (2011)PubMedCentralPubMedCrossRef A.R. Cyr, F.E. Domann, The redox basis of epigenetic modifications: from mechanisms to functional consequences. Antioxid Redox Signal 15, 551–589 (2011)PubMedCentralPubMedCrossRef
60.
go back to reference R.H. Dowen, M. Pelizzola, R.J. Schmitz, R. Lister, J.M. Dowen, J.R. Nery, J.E. Dixon, J.R. Ecker, Widespread dynamic DNA methylation in response to biotic stress. Proc Natl Acad Sci U S A 109, E2183–E2191 (2012)PubMedCentralPubMedCrossRef R.H. Dowen, M. Pelizzola, R.J. Schmitz, R. Lister, J.M. Dowen, J.R. Nery, J.E. Dixon, J.R. Ecker, Widespread dynamic DNA methylation in response to biotic stress. Proc Natl Acad Sci U S A 109, E2183–E2191 (2012)PubMedCentralPubMedCrossRef
61.
go back to reference K. Schrör, Pharmacology and cellular/molecular mechanisms of action of aspirin and non-aspirin NSAIDs in colorectal cancer. Best Pract Res Clin Gastroenterol 25, 473–484 (2011)PubMedCrossRef K. Schrör, Pharmacology and cellular/molecular mechanisms of action of aspirin and non-aspirin NSAIDs in colorectal cancer. Best Pract Res Clin Gastroenterol 25, 473–484 (2011)PubMedCrossRef
62.
go back to reference E.C. Yiannakopoulou, E. Tiligada, Protective effect of salicylates against hydrogen peroxide stress in yeast. J Appl Microbiol 106, 903–908 (2009)PubMedCrossRef E.C. Yiannakopoulou, E. Tiligada, Protective effect of salicylates against hydrogen peroxide stress in yeast. J Appl Microbiol 106, 903–908 (2009)PubMedCrossRef
63.
go back to reference M.C. de Andrés, K. Imagawa, K. Hashimoto, A. Gonzalez, H.I. Roach, M.B. Goldring, R.O. Oreffo, Loss of methylation in CpG sites in the NF-κB enhancer elements of inducible nitric oxide synthase is responsible for gene induction in human articular chondrocytes. Arthritis Rheum 65, 732–742 (2013)PubMedCentralPubMedCrossRef M.C. de Andrés, K. Imagawa, K. Hashimoto, A. Gonzalez, H.I. Roach, M.B. Goldring, R.O. Oreffo, Loss of methylation in CpG sites in the NF-κB enhancer elements of inducible nitric oxide synthase is responsible for gene induction in human articular chondrocytes. Arthritis Rheum 65, 732–742 (2013)PubMedCentralPubMedCrossRef
64.
go back to reference Fata, J.E.; Debnath, S.; Jenkins, E.C. Jr; Fournier, M.V. Nongenomic Mechanisms of PTEN Regulation. Int. J. Cell Biol., 2012, 379685 Fata, J.E.; Debnath, S.; Jenkins, E.C. Jr; Fournier, M.V. Nongenomic Mechanisms of PTEN Regulation. Int. J. Cell Biol., 2012, 379685
65.
go back to reference K.K. Wu, Transcription-based COX-2 inhibition: a therapeutic strategy. Thromb Haemost 96, 417–422 (2006)PubMed K.K. Wu, Transcription-based COX-2 inhibition: a therapeutic strategy. Thromb Haemost 96, 417–422 (2006)PubMed
66.
go back to reference D.A. Dixon, F.F. Blanco, A. Bruno, P. Patrignani, Mechanistic aspects of COX-2 expression in colorectal neoplasia. Recent Results Cancer Res 191, 7–37 (2013)PubMedCentralPubMedCrossRef D.A. Dixon, F.F. Blanco, A. Bruno, P. Patrignani, Mechanistic aspects of COX-2 expression in colorectal neoplasia. Recent Results Cancer Res 191, 7–37 (2013)PubMedCentralPubMedCrossRef
67.
Metadata
Title
Targeting epigenetic mechanisms and microRNAs by aspirin and other non steroidal anti-inflammatory agents - implications for cancer treatment and chemoprevention
Author
Eugenia Yiannakopoulou
Publication date
01-06-2014
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 3/2014
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-014-0175-7

Other articles of this Issue 3/2014

Cellular Oncology 3/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine