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...

Inhibition of NF-κB by Dehydroxymethylepoxyquinomicin Suppresses Invasion and Synergistically Potentiates Temozolomide and γ-Radiation Cytotoxicity in Glioblastoma Cells

DOI: 10.1155/2013/593020

Full-Text   Cite this paper   Add to My Lib

Abstract:

Despite advances in neurosurgery and aggressive treatment with temozolomide (TMZ) and radiation, the overall survival of patients with glioblastoma (GBM) remains poor. Vast evidence has indicated that the nuclear factor NF-κB is constitutively activated in cancer cells, playing key roles in growth and survival. Recently, Dehydroxymethylepoxyquinomicin (DHMEQ) has shown to be a selective NF-κB inhibitor with antiproliferative properties in GBM. In the present study, the ability of DHMEQ to surmount tumor's invasive nature and therapy resistance were further explored. Corroborating results showed that DHMEQ impaired cell growth in dose- and time-dependent manners with G2/M arrest when compared with control. Clonogenicity was also significantly diminished with increased apoptosis, though necrotic cell death was also observed at comparable levels. Notably, migration and invasion were inhibited accordingly with lowered expression of invasion-related genes. Moreover, concurrent combination with TMZ synergistically inhibited cell growth in all cell lines, as determined by proliferation and caspase-3 activation assays, though in those that express O6-methylguanine-DNA methyltransferase, the synergistic effects were schedule dependent. Pretreatment with DHMEQ equally sensitized cells to ionizing radiation. Taken together, our results strengthen the potential usefulness of DHMEQ in future therapeutic strategies for tumors that do not respond to conventional approaches. 1. Introduction Glioblastoma (GBM) is the most aggressive primary brain tumor [1]. Despite improvements in neurosurgery, radiation management, and the advent of temozolomide (TMZ), the outcome of patients remains extremely poor, with a mean life expectancy of approximately one year [2], owing to its ability to infiltrate/invade surrounding tissues and inherent radio- and chemoresistance. Over the past decade, compelling evidence demonstrated that constitutive activation of NF-κB and aberrant regulation of the signaling pathways that control its activity are involved in cancer development and progression, as well as in resistance to therapy in many types of malignancies including GBM [3–6]. Thus, inhibition of the NF-κB pathway seems to be a promising option to improve the efficacy of conventional anticancer therapies. A plethora of NF-κB inhibitors has shown to be effective against various carcinomas and lymphomas (including proteasome inhibitors, IKK inhibitors, and inhibitors of IkB phosphorylation) [7], though, most of them barely discriminate between malignant and normal cells and result in

References

[1]  H. Ohgaki and P. Kleihues, “Genetic pathways to primary and secondary glioblastoma,” American Journal of Pathology, vol. 170, no. 5, pp. 1445–1453, 2007.
[2]  R. Stupp, M. E. Hegi, W. P. Mason, et al., “Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial,” The Lancet Oncology, vol. 10, no. 5, pp. 459–466, 2009.
[3]  S. Nagai, K. Washiyama, M. Kurimoto, A. Takaku, S. Endo, and T. Kumanishi, “Aberrant nuclear factor-κB activity and its participation in the growth of human malignant astrocytoma,” Journal of Neurosurgery, vol. 96, no. 5, pp. 909–917, 2002.
[4]  D. S. Bassères and A. S. Baldwin, “Nuclear factor-κB and inhibitor of κB kinase pathways in oncogenic initiation and progression,” Oncogene, vol. 25, no. 51, pp. 6817–6830, 2006.
[5]  B. Raychaudhuri, Y. Han, T. Lu, and M. A. Vogelbaum, “Aberrant constitutive activation of nuclear factor κB in glioblastoma multiforme drives invasive phenotype,” Journal of Neuro-Oncology, vol. 85, no. 1, pp. 39–47, 2007.
[6]  D. Smith, T. Shimamura, S. Barbera, and B. E. Bejcek, “NF-κB controls growth of glioblastomas/astrocytomas,” Molecular and Cellular Biochemistry, vol. 307, no. 1-2, pp. 141–147, 2008.
[7]  T. D. Gilmore and M. R. Garbati, “Inhibition of NF-κB signaling as a strategy in disease therapy,” Current Topics in Microbiology and Immunology, vol. 349, pp. 245–263, 2011.
[8]  M. Yamamoto, R. Horie, M. Takeiri, I. Kozawa, and K. Umezawa, “Inactivation of NF-κB components by covalent binding of (-)-dehydroxymethylepoxyquinomicin to specific cysteine residues,” Journal of Medicinal Chemistry, vol. 51, no. 18, pp. 5780–5788, 2008.
[9]  K. Umezawa, “Inhibition of tumor growth by NF-κB inhibitors,” Cancer Science, vol. 97, no. 10, pp. 990–995, 2006.
[10]  T. Fukushima, M. Kawaguchi, K. Yorita, et al., “Antitumor effect of dehydroxymethylepoxyquinomicin, a small molecule inhibitor of nuclear factor-κB, on glioblastoma,” Neuro-Oncology, vol. 14, no. 1, pp. 19–28, 2012.
[11]  Y. Suzuki, C. Sugiyama, O. Ohno, et al., “Preparation and biological activities of optically active dehydroxymethylepoxyquinomicin, a novel nuclear factor-κB inhibitor,” Tetrahedran, vol. 60, pp. 7061–7066, 2004.
[12]  N. A. P. Franken, H. M. Rodermond, J. Stap, J. Haveman, and C. van Bree, “Clonogenic assay of cells in vitro,” Nature Protocols, vol. 1, no. 5, pp. 2315–2319, 2006.
[13]  Y. J. Lee and E. Shacter, “Oxidative stress inhibits apoptosis in human lymphoma cells,” Journal of Biological Chemistry, vol. 274, no. 28, pp. 19792–19798, 1999.
[14]  C. C. Liang, A. Y. Park, and J. L. Guan, “In vitro scratch assay: a convenient and inexpensive method for analysis of cell migration in vitro,” Nature Protocols, vol. 2, no. 2, pp. 329–333, 2007.
[15]  B. Pauwels, A. E. C. Korst, C. M. J. De Pooter et al., “Comparison of the sulforhodamine B assay and the clonogenic assay for in vitro chemoradiation studies,” Cancer Chemotherapy and Pharmacology, vol. 51, no. 3, pp. 221–226, 2003.
[16]  Y. Tao, P. Zhang, V. Frascogna et al., “Enhancement of radiation response by inhibition of Aurora-A kinase using siRNA or a selective Aurora kinase inhibitor PHA680632 in p53-deficient cancer cells,” British Journal of Cancer, vol. 97, no. 12, pp. 1664–1672, 2007.
[17]  K. J. Livak and T. D. Schmittgen, “Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method,” Methods, vol. 25, no. 4, pp. 402–408, 2001.
[18]  J. G. Herman, J. R. Graff, S. My?h?nen, B. D. Nelkin, and S. B. Baylin, “Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands,” Proceedings of the National Academy of Sciences of the United States of America, vol. 93, no. 18, pp. 9821–9826, 1996.
[19]  S. J. Clark, J. Harrison, C. L. Paul, and M. Frommer, “High sensitivity mapping of methylated cytosines,” Nucleic Acids Research, vol. 22, no. 15, pp. 2990–2997, 1994.
[20]  D. Beier, S. R?hrl, D. R. Pillai et al., “Temozolomide preferentially depletes cancer stem cells in glioblastoma,” Cancer Research, vol. 68, no. 14, pp. 5706–5715, 2008.
[21]  N. P. Singh, M. T. McCoy, R. R. Tice, and E. L. Schneider, “A simple technique for quantitation of low levels of DNA damage in individual cells,” Experimental Cell Research, vol. 175, no. 1, pp. 184–191, 1988.
[22]  T. C. Chou and P. Talalay, “Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors,” Advances in Enzyme Regulation, vol. 22, no. C, pp. 27–55, 1984.
[23]  B. B. Aggarwal, “Nuclear factor-κB: the enemy within,” Cancer Cell, vol. 6, no. 3, pp. 203–208, 2004.
[24]  P. A. Robe, M. Bentires-Alj, M. Bonif et al., “In vitro and in vivo activity of the nuclear factor-κB inhibitor sulfasalazine in human glioblastomas,” Clinical Cancer Research, vol. 10, no. 16, pp. 5595–5603, 2004.
[25]  K. M. Dhandapani, V. B. Mahesh, and D. W. Brann, “Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFκB transcription factors,” Journal of Neurochemistry, vol. 102, no. 2, pp. 522–538, 2007.
[26]  D. Kesanakurti, G. R. Sareddy, P. P. Babu, and P. B. Kirti, “Mustard NPR1, a mammalian IκB homologue inhibits NF-κB activation in human GBM cell lines,” Biochemical and Biophysical Research Communications, vol. 390, no. 3, pp. 427–433, 2009.
[27]  D. Aguilar-Morante, J. A. Morales-Garcia, M. Sanz-Sancristobal, M. A. Garcia-Cabezas, A. Santos, and A. Perez-Castillo, “Inhibition of glioblastoma growth by the thiadiazolidinone compound TDZD-8,” PLoS One, vol. 5, no. 11, Article ID e13879, 2010.
[28]  Y. Fujiwara, Y. Komohara, T. Ikeda, and M. Takeya, “Corosolic acid inhibits glioblastoma cell proliferation by suppressing the activation of signal transducer and activator of transcription-3 and nuclear factor-kappa B in tumor cells and tumor-associated macrophages,” Cancer Science, vol. 102, no. 1, pp. 206–211, 2011.
[29]  A. Zanotto-Filho, E. Braganhol, R. Schr?der et al., “NFκB inhibitors induce cell death in glioblastomas,” Biochemical Pharmacology, vol. 81, no. 3, pp. 412–424, 2011.
[30]  E. Kikuchi, Y. Horiguchi, J. Nakashima et al., “Suppression of hormone-refractory prostate cancer by a novel nuclear factor κB inhibitor in nude mice,” Cancer Research, vol. 63, no. 1, pp. 107–110, 2003.
[31]  D. V. Starenki, H. Namba, V. A. Saenko et al., “Induction of thyroid cancer cell apoptosis by a novel nuclear factor κB inhibitor, dehydroxymethylepoxyquinomicin,” Clinical Cancer Research, vol. 10, no. 20, pp. 6821–6829, 2004.
[32]  G. Matsumoto, M. Muta, K. Umezawa et al., “Enhancement of the caspase-independent apoptotic sensitivity of pancreatic cancer cells by DHMEQ, an NF-kappaB inhibitor,” International Journal of Oncology, vol. 27, no. 5, pp. 1247–1255, 2005.
[33]  T. Ohsugi, R. Horie, T. Kumasaka et al., “In vivo antitumor activity of the NF-κB inhibitor dehydroxymethylepoxyquinomicin in a mouse model of adult T-cell leukemia,” Carcinogenesis, vol. 26, no. 8, pp. 1382–1388, 2005.
[34]  H. Y. Ruan, M. Masuda, A. Ito et al., “Effects of a novel NF-κB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), on growth, apoptosis, gene expression, and chemosensitivity in head and neck squamous cell carcinoma cell lines,” Head and Neck, vol. 28, no. 2, pp. 158–165, 2006.
[35]  K. Mino, M. Ozaki, K. Nakanishi et al., “Inhibition of nuclear factor-kappaB suppresses peritoneal dissemination of gastric cancer by blocking cancer cell adhesion,” Cancer Science, vol. 102, no. 5, pp. 1052–1058, 2011.
[36]  K. Kodaira, E. Kikuchi, M. Kosugi et al., “Potent cytotoxic effect of a novel nuclear factor-κB inhibitor dehydroxymethylepoxyquinomicin on human bladder cancer cells producing various cytokines,” Urology, vol. 75, no. 4, pp. 805–812, 2010.
[37]  A. Yasuda, S. Kondo, T. Nagumo et al., “Anti-tumor activity of dehydroxymethylepoxyquinomicin against human oral squamous cell carcinoma cell lines in vitro and in vivo,” Oral Oncology, vol. 47, no. 5, pp. 334–339, 2011.
[38]  H. Tatetsu, Y. Okuno, M. Nakamura et al., “Dehydroxymethylepoxyquinomicin, a novel nuclear factor-κB inhibitor, induces apoptosis in multiple myeloma cells in an IkBα-independent manner,” Molecular Cancer Therapeutics, vol. 4, no. 7, pp. 1114–1120, 2005.
[39]  D. Nishimura, H. Ishikawa, K. Matsumoto et al., “DHMEQ, a novel NF-κB inhibitor, induces apoptosis and cell-cycle arrest in human hepatoma cells,” International Journal of Oncology, vol. 29, no. 3, pp. 713–719, 2006.
[40]  N. Kimura, Y. Miyakawa, K. Kohmura, K. Umezawa, Y. Ikeda, and M. Kizaki, “Targeting NF-κB and induction of apoptosis by novel NF-κB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) in Burkitt lymphoma cells,” Leukemia Research, vol. 31, no. 11, pp. 1529–1535, 2007.
[41]  M. M. Chaturvedi, B. Sung, V. R. Yadav, R. Kannappan, and B. B. Aggarwal, “NF-κB addiction and its role in cancer: one size does not fit all,” Oncogene, vol. 30, no. 14, pp. 1615–1630, 2011.
[42]  H. L. Pahl, “Activators and target genes of Rel/NF-κB transcription factors,” Oncogene, vol. 18, no. 49, pp. 6853–6866, 1999.
[43]  F. Wan and M. J. Lenardo, “Specification of DNA binding activity of NF-kappaB proteins,” Cold Spring Harbor Perspectives in Biology, vol. 1, no. 4, Article ID a000067, 2009.
[44]  Z. Jiang, X. Zheng, and K. M. Rich, “Down-regulation of Bcl-2 and Bcl-xL expression with bispecific antisense treatment in glioblastoma cell lines induce cell death,” Journal of Neurochemistry, vol. 84, no. 2, pp. 273–281, 2003.
[45]  S. K. Chintala, J. C. Tonn, and J. S. Rao, “Matrix metalloproteinases and their biological function in human gliomas,” International Journal of Developmental Neuroscience, vol. 17, no. 5-6, pp. 495–502, 1999.
[46]  P. M. Mohan, S. S. Lakka, S. Mohanam et al., “Downregulation of the urokinase-type plasminogen activator receptor through inhibition of translation by antisense oligonucleotide suppresses invasion of human glioblastoma cells,” Clinical and Experimental Metastasis, vol. 17, no. 7, pp. 617–621, 1999.
[47]  V. B. Andela, E. M. Schwarz, J. E. Puzas, R. J. O'Keefe, and R. N. Rosier, “Tumor metastasis and the reciprocal regulation of prometastatic and antimetastatic factors by nuclear factor κB,” Cancer Research, vol. 60, no. 23, pp. 6557–6562, 2000.
[48]  K. Tsunoda, G. Kitange, T. Anda et al., “Expression of the constitutively activated RelA/NF-κB in human astrocytic tumors and the in vitro implication in the regulation of urokinase-type plasminogen activator, migration, and invasion,” Brain Tumor Pathology, vol. 22, no. 2, pp. 79–87, 2005.
[49]  A. Giese, R. Bjerkvig, M. E. Berens, and M. Westphal, “Cost of migration: invasion of malignant gliomas and implications for treatment,” Journal of Clinical Oncology, vol. 21, no. 8, pp. 1624–1636, 2003.
[50]  G. Iacob and E. B. Dinca, “Current data and strategy in glioblastoma multiforme,” Journal of Medicine and Life, vol. 2, no. 4, pp. 386–393, 2009.
[51]  C. Y. Wang, J. C. Cusack Jr., R. Liu, and A. S. Baldwin Jr., “Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-κB,” Nature Medicine, vol. 5, no. 4, pp. 412–417, 1999.
[52]  M. Bignami, M. O'Driscoll, G. Aquilina, and P. Karran, “Unmasking a killer: DNA O6-methylguanine and the cytotoxicity of methylating agents,” Mutation Research, vol. 462, no. 2-3, pp. 71–82, 2000.
[53]  M. Belanich, M. Pastor, T. Randall et al., “Retrospective study of the correlation between the DNA repair protein alkyltransferase and survival of brain tumor patients treated with carmustine,” Cancer Research, vol. 56, no. 4, pp. 783–788, 1996.
[54]  N. Shah, B. Lin, Z. Sibenaller et al., “Comprehensive analysis of MGMT promoter methylation: correlation with MGMT expression and clinical response in GBM,” PLoS One, vol. 6, no. 1, Article ID e16146, 2011.
[55]  P. Poma, M. Notarbartolo, M. Labbozzetta et al., “Antitumor effects of the novel NF-κB inhibitor dehydroxymethylepoxyquinomicin on human hepatic cancer cells: analysis of synergy with cisplatin and of possible correlation with inhibition of pro-survival genes and IL-6 production,” International Journal of Oncology, vol. 28, no. 4, pp. 923–930, 2006.
[56]  Z. Meng, N. Mitsutake, M. Nakashima et al., “Dehydroxymethylepoxyquinomicin, a novel nuclear factor-κB inhibitor, enhances antitumor activity of taxanes in anaplastic thyroid cancer cells,” Endocrinology, vol. 149, no. 11, pp. 5357–5365, 2008.
[57]  R. Horie, M. Watanabe, T. Okamura et al., “DHMEQ, a new NF-κB inhibitor, induces apoptosis and enhances fludarabine effects on chronic lymphocytic leukemia cells,” Leukemia, vol. 20, no. 5, pp. 800–806, 2006.
[58]  A. R. Jazirehi, M. I. Vega, and B. Bonavida, “Development of rituximab-resistant lymphoma clones with altered cell signaling and cross-resistance to chemotherapy,” Cancer Research, vol. 67, no. 3, pp. 1270–1281, 2007.
[59]  N. Lampiasi, A. Azzolina, K. Umezawa, G. Montalto, J. A. McCubrey, and M. Cervello, “The novel NF-κB inhibitor DHMEQ synergizes with celecoxib to exert antitumor effects on human liver cancer cells by a ROS-dependent mechanism,” Cancer Letters, vol. 322, no. 1, pp. 35–44, 2012.
[60]  M. Bredel, C. Bredel, D. Juric et al., “Tumor necrosis factor-α-induced protein 3 as a putative regulator of nuclear factor-κB-mediated resistance to O6-alkylating agents in human glioblastomas,” Journal of Clinical Oncology, vol. 24, no. 2, pp. 274–287, 2006.
[61]  I. Lavon, D. Fuchs, D. Zrihan et al., “Novel mechanism whereby nuclear factor κB mediates DNA damage repair through regulation of O6-methylguanine-DNA-methyltransferase,” Cancer Research, vol. 67, no. 18, pp. 8952–8959, 2007.
[62]  H. S. Friedman, T. Kerby, and H. Calvert, “Temozolomide and treatment of malignant glioma,” Clinical Cancer Research, vol. 6, no. 7, pp. 2585–2597, 2000.
[63]  N. Yamagishi, J. Miyakoshi, and H. Takebe, “Enhanced radiosensitivity by inhibition of nuclear factor κB activation in human malignant glioma cells,” International Journal of Radiation Biology, vol. 72, no. 2, pp. 157–162, 1997.
[64]  U. Raju, G. J. Gumin, and P. J. Tofilon, “NF kappa B activity and target gene expression in the rat brain after one and two exposures to ionizing radiation,” Radiation Oncology Investigations, vol. 7, no. 3, pp. 145–152, 1999.
[65]  F. Pajonk, K. Pajonk, and W. H. McBride, “Apoptosis and radiosensitization of Hodgkin cells by proteasome inhibition,” International Journal of Radiation Oncology, Biology, Physics, vol. 47, no. 4, pp. 1025–1032, 2000.
[66]  A. S. Baldwin, “Control of oncogenesis and cancer therapy resistance by the transcription factor NF-κB,” Journal of Clinical Investigation, vol. 107, no. 3, pp. 241–246, 2001.
[67]  H. K. G. Shu, M. M. Kim, P. Chen, F. Furman, C. M. Julin, and M. A. Israel, “The intrinsic radioresistance of glioblastoma-derived cell lines is associated with a failure of p53 to induce p21BAX expression,” Proceedings of the National Academy of Sciences of the United States of America, vol. 95, no. 24, pp. 14453–14458, 1998.
[68]  G. Afshar, N. Jelluma, X. Yang et al., “Radiation-induced caspase-8 mediates p53-independent apoptosis in glioma cells,” Cancer Research, vol. 66, no. 8, pp. 4223–4232, 2006.
[69]  Y. Hirose, M. S. Berger, and R. O. Pieper, “p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells,” Cancer Research, vol. 61, no. 5, pp. 1957–1963, 2001.
[70]  H. Ohgaki, P. Dessen, B. Jourde et al., “Genetic pathways to glioblastoma: a population-based study,” Cancer Research, vol. 64, no. 19, pp. 6892–6899, 2004.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413