Skip to main content
Top
Published in: Cellular Oncology 6/2015

01-12-2015 | Original Paper

Role of JNK and NF-κB in mediating the effect of combretastatin A-4 and brimamin on endothelial and carcinoma cells

Authors: Katharina Mahal, Aamir Ahmad, Seema Sethi, Marcus Resch, Ralf Ficner, Fazlul H. Sarkar, Rainer Schobert, Bernhard Biersack

Published in: Cellular Oncology | Issue 6/2015

Login to get access

Abstract

Purpose

The 4,5-diarylimidazole brimamin is an analog of the natural vascular-disrupting agent combretastatin A-4 (CA-4) with improved water solubility, tolerance by animals and efficacy in multidrug-resistant tumors. Here, we aimed at identifying the major mechanisms underlying the in vitro and in vivo actions of brimamin on endothelial and carcinoma cells, including vascularization.

Methods

The contribution of specific signaling kinases to the effects of brimamin on cytoskeleton organization and the viability and differentiation of endothelial cells was assessed by MTT and tube formation assays in the presence or absence of specific kinase inhibitors. Changes in DNA affinity and expression of NF-κB in endothelial and carcinoma-derived cells and their solid tumors (xenografts) treated with brimamin were ascertained by electrophoretic mobility shift assays and Western blotting. The anti-vascular effect of brimamin in solid tumors was verified by CD31 immunostaining.

Results

We found that brimamin can inhibit tubulin polymerization and cause a reorganization of F-actin in Ea.hy926 endothelial cells. Its inhibitory effect on tube formation was found to depend on functional Rho kinase and JNK. JNK inhibition was found to suppress the induction of endothelial cell apoptosis by brimamin. In CA-4-refractory human BxPC-3 pancreas carcinoma-derived and triple-negative MDA-MB-231 breast carcinoma-derived cells brimamin was found to inhibit growth and to induce apoptosis at low nanomolar concentrations by blocking NF-κB activation in a dose-dependent manner. Brimamin was also found to reduce the in vivo growth rate and vascularization of MDA-MB-231 xenografts in mice. Residual tumor cells of these treated xenografts showed a relatively low expression of the p65 subunit of NF-κB.

Conclusions

Our data indicate that cellular JNK and Rho kinase activities are crucial for the cytotoxic and cytoskeleton reorganizing effects of brimamin on endothelial cells. In addition, we found that in resistant carcinoma cells and xenografts brimamin can induce down-regulation of anti-apoptotic NF-κB expression and signaling. Its chemical properties and efficacy against clinically relevant cancer entities make brimamin a promising candidate vascular-disrupting agent.
Appendix
Available only for authorised users
Literature
1.
go back to reference G. Pettit, S. Singh, E. Hamel, C. Lin, D. Alberts, D. Garcia-Kendall, Isolation and structure of the strong cell growth and tubulin inhibitor combretastatin A-4. Experientia 45, 209–211 (1989)CrossRefPubMed G. Pettit, S. Singh, E. Hamel, C. Lin, D. Alberts, D. Garcia-Kendall, Isolation and structure of the strong cell growth and tubulin inhibitor combretastatin A-4. Experientia 45, 209–211 (1989)CrossRefPubMed
2.
go back to reference C.J. Mooney, G. Nagaiah, P. Fu, J.K. Wasman, M.M. Cooney, P.S. Savvides, J.A. Bokar, A. Dowlati, D. Wang, S.S. Agarwala, A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid 19, 233–240 (2009)PubMedCentralCrossRefPubMed C.J. Mooney, G. Nagaiah, P. Fu, J.K. Wasman, M.M. Cooney, P.S. Savvides, J.A. Bokar, A. Dowlati, D. Wang, S.S. Agarwala, A phase II trial of fosbretabulin in advanced anaplastic thyroid carcinoma and correlation of baseline serum-soluble intracellular adhesion molecule-1 with outcome. Thyroid 19, 233–240 (2009)PubMedCentralCrossRefPubMed
3.
go back to reference G.J. Rustin, G. Shreeves, P.D. Nathan, A. Gaya, T.S. Ganesan, D. Wang, J. Boxall, L. Poupard, D.J. Chaplin, M.R.L. Stratford, J. Balkissoon, M. Zweifel, A Phase Ib trial of CA4P (combretastatin A-4 phosphate), carboplatin, and paclitaxel in patients with advanced cancer. Br. J. Cancer 102, 1355–1360 (2010)PubMedCentralCrossRefPubMed G.J. Rustin, G. Shreeves, P.D. Nathan, A. Gaya, T.S. Ganesan, D. Wang, J. Boxall, L. Poupard, D.J. Chaplin, M.R.L. Stratford, J. Balkissoon, M. Zweifel, A Phase Ib trial of CA4P (combretastatin A-4 phosphate), carboplatin, and paclitaxel in patients with advanced cancer. Br. J. Cancer 102, 1355–1360 (2010)PubMedCentralCrossRefPubMed
4.
go back to reference G.C. Tron, T. Pirali, G. Sorba, F. Pagliai, S. Busacca, A.A. Genazzani, Medicinal chemistry of combretastatin A4: present and future directions. J. Med. Chem. 49, 3033–3044 (2006)CrossRefPubMed G.C. Tron, T. Pirali, G. Sorba, F. Pagliai, S. Busacca, A.A. Genazzani, Medicinal chemistry of combretastatin A4: present and future directions. J. Med. Chem. 49, 3033–3044 (2006)CrossRefPubMed
5.
go back to reference S.E. Holwell, P.A. Cooper, M.J. Thompson, G.R. Pettit, L.W. Lippert 3rd, S.W. Martin, M.C. Bibby, Anti-tumor and anti-vascular effects of the novel tubulin-binding agent combretastatin A-1 phosphate. Anticancer Res. 22, 3933–3940 (2002)PubMed S.E. Holwell, P.A. Cooper, M.J. Thompson, G.R. Pettit, L.W. Lippert 3rd, S.W. Martin, M.C. Bibby, Anti-tumor and anti-vascular effects of the novel tubulin-binding agent combretastatin A-1 phosphate. Anticancer Res. 22, 3933–3940 (2002)PubMed
6.
go back to reference L.K. Folkes, M. Christlieb, E. Madej, M.R.L. Stratford, P. Wardman, Oxidative metabolism of combretastatin A-1 produces quinone intermediates with the potential to bind to nucleophiles and to enhance oxidative stress via free radicals. Chem. Res. Toxicol. 20, 1885–1894 (2007)CrossRefPubMed L.K. Folkes, M. Christlieb, E. Madej, M.R.L. Stratford, P. Wardman, Oxidative metabolism of combretastatin A-1 produces quinone intermediates with the potential to bind to nucleophiles and to enhance oxidative stress via free radicals. Chem. Res. Toxicol. 20, 1885–1894 (2007)CrossRefPubMed
7.
go back to reference G.R. Pettit, M.R. Rhodes, D.L. Herald, D.J. Chaplin, M.R. Stratford, E. Hamel, R.K. Pettit, J.C. Chapuis, D. Oliva, Antineoplastic agents 393. Synthesis of the trans-isomer of combretastatin A-4 prodrug. Anticancer Drug Des. 13, 981–993 (1998)PubMed G.R. Pettit, M.R. Rhodes, D.L. Herald, D.J. Chaplin, M.R. Stratford, E. Hamel, R.K. Pettit, J.C. Chapuis, D. Oliva, Antineoplastic agents 393. Synthesis of the trans-isomer of combretastatin A-4 prodrug. Anticancer Drug Des. 13, 981–993 (1998)PubMed
8.
go back to reference G.R. Pettit, B.E. Toki, D.L. Herald, M.R. Boyd, E. Hamel, R.K. Pettit, J.C. Chapuis, Antineoplastic agents. 410. Asymmetric hydroxylation of trans-combretastatin A-4. J. Med. Chem. 42, 1459–1465 (1999)CrossRefPubMed G.R. Pettit, B.E. Toki, D.L. Herald, M.R. Boyd, E. Hamel, R.K. Pettit, J.C. Chapuis, Antineoplastic agents. 410. Asymmetric hydroxylation of trans-combretastatin A-4. J. Med. Chem. 42, 1459–1465 (1999)CrossRefPubMed
9.
go back to reference L. Wang, K.W. Woods, Q. Li, K.J. Barr, R.W. McCroskey, S.M. Hannick, L. Gherke, R.B. Credo, Y.-H. Hui, K. Marsh, R. Warner, J.Y. Lee, N. Zielinski-Mozng, D. Frost, S.H. Rosenberg, H.L. Sham, Potent, orally active heterocycle-based combretastatin A-4 analogues: synthesis, structure − activity relationship, pharmacokinetics, and in vivo antitumor activity evaluation. J. Med. Chem. 45, 1697–1711 (2002)CrossRefPubMed L. Wang, K.W. Woods, Q. Li, K.J. Barr, R.W. McCroskey, S.M. Hannick, L. Gherke, R.B. Credo, Y.-H. Hui, K. Marsh, R. Warner, J.Y. Lee, N. Zielinski-Mozng, D. Frost, S.H. Rosenberg, H.L. Sham, Potent, orally active heterocycle-based combretastatin A-4 analogues: synthesis, structure − activity relationship, pharmacokinetics, and in vivo antitumor activity evaluation. J. Med. Chem. 45, 1697–1711 (2002)CrossRefPubMed
10.
go back to reference R. Schobert, B. Biersack, A. Dietrich, K. Effenberger, S. Knauer, T. Mueller, 4-(3-Halo/amino-4,5-dimethoxyphenyl)-5-aryloxazoles and -N-methylimidazoles that are cytotoxic against combretastatin A resistant tumor cells and vascular disrupting in a cisplatin resistant germ cell tumor model. J. Med. Chem. 53, 6595–6602 (2010)CrossRefPubMed R. Schobert, B. Biersack, A. Dietrich, K. Effenberger, S. Knauer, T. Mueller, 4-(3-Halo/amino-4,5-dimethoxyphenyl)-5-aryloxazoles and -N-methylimidazoles that are cytotoxic against combretastatin A resistant tumor cells and vascular disrupting in a cisplatin resistant germ cell tumor model. J. Med. Chem. 53, 6595–6602 (2010)CrossRefPubMed
11.
go back to reference K. Mahal, B. Biersack, H. Caysa, R. Schobert, T. Mueller, Combretastatin A-4 derived imidazoles show cytotoxic, antivascular, and antimetastatic effects based on cytoskeletal reorganisation. Investig. New Drugs 33, 541–554 (2015)CrossRef K. Mahal, B. Biersack, H. Caysa, R. Schobert, T. Mueller, Combretastatin A-4 derived imidazoles show cytotoxic, antivascular, and antimetastatic effects based on cytoskeletal reorganisation. Investig. New Drugs 33, 541–554 (2015)CrossRef
12.
go back to reference C. Kanthou, The tumor vascular targeting agent combretastatin A-4-phosphate induces reorganization of the actin cytoskeleton and early membrane blebbing in human endothelial cells. Blood 99, 2060–2069 (2002)CrossRefPubMed C. Kanthou, The tumor vascular targeting agent combretastatin A-4-phosphate induces reorganization of the actin cytoskeleton and early membrane blebbing in human endothelial cells. Blood 99, 2060–2069 (2002)CrossRefPubMed
13.
go back to reference B.B. Aggarwal, S. Shishodia, S.K. Sandur, M.K. Pandey, G. Sethi, Inflammation and cancer: how hot is the link? Biochem. Pharmacol. 72, 1605–1621 (2006)CrossRefPubMed B.B. Aggarwal, S. Shishodia, S.K. Sandur, M.K. Pandey, G. Sethi, Inflammation and cancer: how hot is the link? Biochem. Pharmacol. 72, 1605–1621 (2006)CrossRefPubMed
14.
go back to reference A. Ahmad, S. Banerjee, Z. Wang, D. Kong, F.H. Sarkar, Plumbagin-induced apoptosis of human breast cancer cells is mediated by inactivation of NF-κB and Bcl-2. J. Cell. Biochem. 105, 1461–1471 (2008)CrossRefPubMed A. Ahmad, S. Banerjee, Z. Wang, D. Kong, F.H. Sarkar, Plumbagin-induced apoptosis of human breast cancer cells is mediated by inactivation of NF-κB and Bcl-2. J. Cell. Biochem. 105, 1461–1471 (2008)CrossRefPubMed
15.
go back to reference S. Ali, A. Ahmad, S. Banerjee, S. Padhye, K. Dominiak, J.M. Schaffert, Z. Wang, P.A. Philip, F.H. Sarkar, Gemcitabine sensitivity can be induced in pancreatic cancer cells through modulation of miR-200 and miR-21 expression by curcumin or its analogue CDF. Cancer Res. 70, 3606–3617 (2010)PubMedCentralCrossRefPubMed S. Ali, A. Ahmad, S. Banerjee, S. Padhye, K. Dominiak, J.M. Schaffert, Z. Wang, P.A. Philip, F.H. Sarkar, Gemcitabine sensitivity can be induced in pancreatic cancer cells through modulation of miR-200 and miR-21 expression by curcumin or its analogue CDF. Cancer Res. 70, 3606–3617 (2010)PubMedCentralCrossRefPubMed
16.
go back to reference Y. He, H. Xu, L. Liang, Z. Zhan, X. Yang, X. Yu, Y. Ye, L. Sun, Antiinflammatory effect of Rho kinase blockade via inhibition of NF-kappaB activation in rheumatoid arthritis. Arthritis Rheum. 58, 3366–3376 (2008)CrossRefPubMed Y. He, H. Xu, L. Liang, Z. Zhan, X. Yang, X. Yu, Y. Ye, L. Sun, Antiinflammatory effect of Rho kinase blockade via inhibition of NF-kappaB activation in rheumatoid arthritis. Arthritis Rheum. 58, 3366–3376 (2008)CrossRefPubMed
17.
go back to reference H. Shimada, L.E. Rajagopalan, Rho kinase-2 activation in human endothelial cells drives lysophosphatidic acid-mediated expression of cell adhesion molecules via NF-kappaB p65. J. Biol. Chem. 285, 12536–12542 (2010)PubMedCentralCrossRefPubMed H. Shimada, L.E. Rajagopalan, Rho kinase-2 activation in human endothelial cells drives lysophosphatidic acid-mediated expression of cell adhesion molecules via NF-kappaB p65. J. Biol. Chem. 285, 12536–12542 (2010)PubMedCentralCrossRefPubMed
18.
go back to reference S. Shimizu, M. Tahara, S. Ogata, K. Hashimoto, K. Morishige, K. Tasaka, Y. Murata, Involvement of nuclear factor-kappaB activation through RhoA/Rho-kinase pathway in LPS-induced IL-8 production in human cervical stromal cells. Mol. Hum. Reprod. 13, 181–187 (2007)CrossRefPubMed S. Shimizu, M. Tahara, S. Ogata, K. Hashimoto, K. Morishige, K. Tasaka, Y. Murata, Involvement of nuclear factor-kappaB activation through RhoA/Rho-kinase pathway in LPS-induced IL-8 production in human cervical stromal cells. Mol. Hum. Reprod. 13, 181–187 (2007)CrossRefPubMed
19.
go back to reference G.G. Mackenzie, C.L. Keen, P.I. Oteiza, Microtubules are required for NF-kappaB nuclear translocation in neuroblastoma IMR-32 cells: modulation by zinc. J. Neurochem. 99, 402–415 (2006)CrossRefPubMed G.G. Mackenzie, C.L. Keen, P.I. Oteiza, Microtubules are required for NF-kappaB nuclear translocation in neuroblastoma IMR-32 cells: modulation by zinc. J. Neurochem. 99, 402–415 (2006)CrossRefPubMed
20.
go back to reference V. Bourgarel-Rey, S. Vallee, O. Rimet, S. Champion, D. Braguer, A. Desobry, C. Briand, Y. Barra, Involvement of nuclear factor kappaB in c-Myc induction by tubulin polymerization inhibitors. Mol. Pharmacol. 59, 1165–1170 (2001)PubMed V. Bourgarel-Rey, S. Vallee, O. Rimet, S. Champion, D. Braguer, A. Desobry, C. Briand, Y. Barra, Involvement of nuclear factor kappaB in c-Myc induction by tubulin polymerization inhibitors. Mol. Pharmacol. 59, 1165–1170 (2001)PubMed
21.
go back to reference T.F. Hansen, B.S. Nielsen, A. Jakobsen, F.B. Sorensen, Visualising and quantifying angiogenesis in metastatic colorectal cancer. A comparison of methods and their predictive value for chemotherapy response. Cell. Oncol. 36, 341–350 (2013)CrossRef T.F. Hansen, B.S. Nielsen, A. Jakobsen, F.B. Sorensen, Visualising and quantifying angiogenesis in metastatic colorectal cancer. A comparison of methods and their predictive value for chemotherapy response. Cell. Oncol. 36, 341–350 (2013)CrossRef
22.
go back to reference B. Pula, M. Olbromski, A. Wojnar, A. Gomulkiewicz, W. Witkiewicz, M. Ugorski, P. Dziegiel, M. Podhorska-Okolow, Impact of SOX18 expression in cancer cells and vessels on the outcome of invasive ductal breast carcinoma. Cell. Oncol. 36, 469–483 (2013)CrossRef B. Pula, M. Olbromski, A. Wojnar, A. Gomulkiewicz, W. Witkiewicz, M. Ugorski, P. Dziegiel, M. Podhorska-Okolow, Impact of SOX18 expression in cancer cells and vessels on the outcome of invasive ductal breast carcinoma. Cell. Oncol. 36, 469–483 (2013)CrossRef
23.
go back to reference A.W. Schüttelkopf, D.M.F. van Aalten, PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60, 1355–1363 (2004)CrossRefPubMed A.W. Schüttelkopf, D.M.F. van Aalten, PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60, 1355–1363 (2004)CrossRefPubMed
24.
go back to reference O. Trott, A.J. Olson, AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455–461 (2009) O. Trott, A.J. Olson, AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 31, 455–461 (2009)
25.
go back to reference J. Gasteiger, M. Marsili, Iterative partial equalization of orbital electronegativity – a rapid access to atomic charges. Tetrahedron 36, 3219–3228 (1980)CrossRef J. Gasteiger, M. Marsili, Iterative partial equalization of orbital electronegativity – a rapid access to atomic charges. Tetrahedron 36, 3219–3228 (1980)CrossRef
26.
go back to reference G.L. Warren, C.W. Andrews, A.-M. Capelli, B. Clarke, J. LaLonde, M.H. Lambert, M. Lindvall, N. Nevins, S.F. Semus, S. Senger, G. Tedesco, I.D. Wall, J.M. Woolven, C.E. Peishoff, M.S. Head, A critical assessment of docking programs and scoring functions. J. Med. Chem. 49, 5912–5931 (2006)CrossRefPubMed G.L. Warren, C.W. Andrews, A.-M. Capelli, B. Clarke, J. LaLonde, M.H. Lambert, M. Lindvall, N. Nevins, S.F. Semus, S. Senger, G. Tedesco, I.D. Wall, J.M. Woolven, C.E. Peishoff, M.S. Head, A critical assessment of docking programs and scoring functions. J. Med. Chem. 49, 5912–5931 (2006)CrossRefPubMed
27.
go back to reference W. DeLano, The PyMOL molecular graphics system (DeLano Sci, LLC San Carlos CA, 2003) W. DeLano, The PyMOL molecular graphics system (DeLano Sci, LLC San Carlos CA, 2003)
28.
go back to reference K. Mahal, S. Schruefer, G. Steinemann, F. Rausch, R. Schobert, M. Höpfner, B. Biersack, Biological evaluation of 4,5-diarylimidazoles with hydroxamic acid appendages as novel dual mode anticancer agents. Cancer Chemother. Pharmacol. 75, 691–700 (2015)CrossRefPubMed K. Mahal, S. Schruefer, G. Steinemann, F. Rausch, R. Schobert, M. Höpfner, B. Biersack, Biological evaluation of 4,5-diarylimidazoles with hydroxamic acid appendages as novel dual mode anticancer agents. Cancer Chemother. Pharmacol. 75, 691–700 (2015)CrossRefPubMed
29.
go back to reference K. Mahal, M. Resch, R. Ficner, R. Schobert, B. Biersack, T. Mueller, Effects of the tumor-vasculature-disrupting agent verubulin and two heteroaryl analogues on cancer cells, endothelial cells, and blood vessels. ChemMedChem 9, 847–854 (2014)CrossRefPubMed K. Mahal, M. Resch, R. Ficner, R. Schobert, B. Biersack, T. Mueller, Effects of the tumor-vasculature-disrupting agent verubulin and two heteroaryl analogues on cancer cells, endothelial cells, and blood vessels. ChemMedChem 9, 847–854 (2014)CrossRefPubMed
30.
go back to reference G.M. Tozer, C. Kanthou, B.C. Baguley, Disrupting tumour blood vessels. Nat. Rev. Cancer 5, 423–435 (2005)CrossRefPubMed G.M. Tozer, C. Kanthou, B.C. Baguley, Disrupting tumour blood vessels. Nat. Rev. Cancer 5, 423–435 (2005)CrossRefPubMed
31.
go back to reference B. Biersack, K. Effenberger, R. Schobert, M. Ocker, Oxazole-bridged combretastatin a analogues with improved anticancer properties. ChemMedChem 5, 420–427 (2010)CrossRefPubMed B. Biersack, K. Effenberger, R. Schobert, M. Ocker, Oxazole-bridged combretastatin a analogues with improved anticancer properties. ChemMedChem 5, 420–427 (2010)CrossRefPubMed
32.
go back to reference S. Ali, Simultaneous targeting of the epidermal growth factor receptor and cyclooxygenase-2 pathways for pancreatic cancer therapy. Mol. Cancer Ther. 4, 1943–1951 (2005)CrossRefPubMed S. Ali, Simultaneous targeting of the epidermal growth factor receptor and cyclooxygenase-2 pathways for pancreatic cancer therapy. Mol. Cancer Ther. 4, 1943–1951 (2005)CrossRefPubMed
33.
go back to reference R.B.G. Ravelli, B. Gigant, P.A. Curmi, I. Jourdain, S. Lachkar, A. Sobel, M. Knossow, Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature 428, 198–202 (2004)CrossRefPubMed R.B.G. Ravelli, B. Gigant, P.A. Curmi, I. Jourdain, S. Lachkar, A. Sobel, M. Knossow, Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature 428, 198–202 (2004)CrossRefPubMed
34.
go back to reference M. Botta, S. Forli, M. Magnani, F. Manetti, Molecular modeling approaches to study the binding mode on tubulin of microtubule destabilizing and stabilizing agents. Top. Curr. Chem. 286, 279–328 (2009)CrossRefPubMed M. Botta, S. Forli, M. Magnani, F. Manetti, Molecular modeling approaches to study the binding mode on tubulin of microtubule destabilizing and stabilizing agents. Top. Curr. Chem. 286, 279–328 (2009)CrossRefPubMed
35.
go back to reference C. Kanthou, G.M. Tozer, Tumour targeting by microtubule-depolymerising vascular disrupting agents. Expert Opin. Ther. Targets 11, 1443–1457 (2007)CrossRefPubMed C. Kanthou, G.M. Tozer, Tumour targeting by microtubule-depolymerising vascular disrupting agents. Expert Opin. Ther. Targets 11, 1443–1457 (2007)CrossRefPubMed
36.
go back to reference H. Quan, Y. Xu, L. Lou, p38 MAPK, but not ERK1/2, is critically involved in the cytotoxicity of the novel vascular disrupting agent combretastatin A4. Int. J. Cancer 122, 1730–1737 (2007)CrossRef H. Quan, Y. Xu, L. Lou, p38 MAPK, but not ERK1/2, is critically involved in the cytotoxicity of the novel vascular disrupting agent combretastatin A4. Int. J. Cancer 122, 1730–1737 (2007)CrossRef
37.
go back to reference M. Fan, L. Du, A. Stone, K. Gilbert, T. Chambers, Modulation of mitogen-activated protein kinases and phosphorylation of Bcl-2 by vinblastine represent persistent forms of normal fluctuations at G2-M. Cancer Res. 60, 6403–6407 (2000)PubMed M. Fan, L. Du, A. Stone, K. Gilbert, T. Chambers, Modulation of mitogen-activated protein kinases and phosphorylation of Bcl-2 by vinblastine represent persistent forms of normal fluctuations at G2-M. Cancer Res. 60, 6403–6407 (2000)PubMed
38.
go back to reference C. Kanthou, G.M. Tozer, Microtubule depolymerizing vascular disrupting agents: novel therapeutic agents for oncology and other pathologies. Int. J. Exp. Pathol. 90, 284–294 (2009)PubMedCentralCrossRefPubMed C. Kanthou, G.M. Tozer, Microtubule depolymerizing vascular disrupting agents: novel therapeutic agents for oncology and other pathologies. Int. J. Exp. Pathol. 90, 284–294 (2009)PubMedCentralCrossRefPubMed
39.
go back to reference M. Chrzanowska-Wodnicka, K. Burridge, Rho-stimulated contractility drives the formation of stress fibers and focal adhesions. J. Cell Biol. 133, 1403–1415 (1996)CrossRefPubMed M. Chrzanowska-Wodnicka, K. Burridge, Rho-stimulated contractility drives the formation of stress fibers and focal adhesions. J. Cell Biol. 133, 1403–1415 (1996)CrossRefPubMed
40.
go back to reference L.J. Williams, D. Mukherjee, M. Fisher, C.C. Reyes-Aldasoro, S. Akerman, C. Kanthou, G.M. Tozer, An in vivo role for Rho kinase activation in the tumour vascular disrupting activity of combretastatin A-4 3-O-phosphate: Rho kinase and tumour vascular targeting. Br. J. Pharmacol. 171, 4902–4913 (2014)PubMedCentralCrossRefPubMed L.J. Williams, D. Mukherjee, M. Fisher, C.C. Reyes-Aldasoro, S. Akerman, C. Kanthou, G.M. Tozer, An in vivo role for Rho kinase activation in the tumour vascular disrupting activity of combretastatin A-4 3-O-phosphate: Rho kinase and tumour vascular targeting. Br. J. Pharmacol. 171, 4902–4913 (2014)PubMedCentralCrossRefPubMed
41.
go back to reference A. Arlt, A. Gehrz, S. Müerköster, J. Vorndamm, M.-L. Kruse, U.R. Fölsch, H. Schäfer, Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene 22, 3243–3251 (2003)CrossRefPubMed A. Arlt, A. Gehrz, S. Müerköster, J. Vorndamm, M.-L. Kruse, U.R. Fölsch, H. Schäfer, Role of NF-kappaB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene 22, 3243–3251 (2003)CrossRefPubMed
42.
go back to reference M. Graupera, J. Guillermet-Guibert, L.C. Foukas, L.-K. Phng, R.J. Cain, A. Salpekar, W. Pearce, S. Meek, J. Millan, P.R. Cutillas, A.J.H. Smith, A.J. Ridley, C. Ruhrberg, H. Gerhardt, B. Vanhaesebroeck, Angiogenesis selectively requires the p110α isoform of PI3K to control endothelial cell migration. Nature 453, 662–666 (2008)CrossRefPubMed M. Graupera, J. Guillermet-Guibert, L.C. Foukas, L.-K. Phng, R.J. Cain, A. Salpekar, W. Pearce, S. Meek, J. Millan, P.R. Cutillas, A.J.H. Smith, A.J. Ridley, C. Ruhrberg, H. Gerhardt, B. Vanhaesebroeck, Angiogenesis selectively requires the p110α isoform of PI3K to control endothelial cell migration. Nature 453, 662–666 (2008)CrossRefPubMed
44.
go back to reference G. Rajashekhar, M. Kamocka, A. Marin, M.A. Suckow, W.R. Wolter, S. Badve, A.R. Sanjeevaiah, K. Pumiglia, E. Rosen, M. Clauss, Pro-inflammatory angiogenesis is mediated by p38 MAP kinase. J. Cell. Physiol. 226, 800–808 (2011)CrossRefPubMed G. Rajashekhar, M. Kamocka, A. Marin, M.A. Suckow, W.R. Wolter, S. Badve, A.R. Sanjeevaiah, K. Pumiglia, E. Rosen, M. Clauss, Pro-inflammatory angiogenesis is mediated by p38 MAP kinase. J. Cell. Physiol. 226, 800–808 (2011)CrossRefPubMed
45.
go back to reference M. Kelkel, C. Cerella, F. Mack, T. Schneider, C. Jacob, M. Schumacher, M. Dicato, M. Diederich, ROS-independent JNK activation and multisite phosphorylation of Bcl-2 link diallyl tetrasulfide-induced mitotic arrest to apoptosis. Carcinogenesis 33, 2162–2171 (2012)CrossRefPubMed M. Kelkel, C. Cerella, F. Mack, T. Schneider, C. Jacob, M. Schumacher, M. Dicato, M. Diederich, ROS-independent JNK activation and multisite phosphorylation of Bcl-2 link diallyl tetrasulfide-induced mitotic arrest to apoptosis. Carcinogenesis 33, 2162–2171 (2012)CrossRefPubMed
46.
go back to reference J.D. Orth, A. Loewer, G. Lahav, T.J. Mitchison, Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol. Biol. Cell 23, 567–576 (2012)PubMedCentralCrossRefPubMed J.D. Orth, A. Loewer, G. Lahav, T.J. Mitchison, Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol. Biol. Cell 23, 567–576 (2012)PubMedCentralCrossRefPubMed
47.
go back to reference S.P. Tabruyn, A.W. Griffioen, A new role for NF-kappaB in angiogenesis inhibition. Cell Death Differ. 14, 1393–1397 (2007)CrossRefPubMed S.P. Tabruyn, A.W. Griffioen, A new role for NF-kappaB in angiogenesis inhibition. Cell Death Differ. 14, 1393–1397 (2007)CrossRefPubMed
48.
go back to reference S.P. Tabruyn, S. Memet, P. Ave, C. Verhaeghe, K.H. Mayo, I. Struman, J.A. Martial, A.W. Griffioen, NF-kappaB activation in endothelial cells is critical for the activity of angiostatic agents. Mol. Cancer Ther. 8, 2645–2654 (2009)CrossRefPubMed S.P. Tabruyn, S. Memet, P. Ave, C. Verhaeghe, K.H. Mayo, I. Struman, J.A. Martial, A.W. Griffioen, NF-kappaB activation in endothelial cells is critical for the activity of angiostatic agents. Mol. Cancer Ther. 8, 2645–2654 (2009)CrossRefPubMed
49.
go back to reference M. Carr, L.M. Greene, A.J.S. Knox, D.G. Lloyd, D.M. Zisterer, M.J. Meegan, Lead identification of conformationally restricted β-lactam type combretastatin analogues: synthesis, antiproliferative activity and tubulin targeting effects. Eur. J. Med. Chem. 45, 5752–5766 (2010)CrossRefPubMed M. Carr, L.M. Greene, A.J.S. Knox, D.G. Lloyd, D.M. Zisterer, M.J. Meegan, Lead identification of conformationally restricted β-lactam type combretastatin analogues: synthesis, antiproliferative activity and tubulin targeting effects. Eur. J. Med. Chem. 45, 5752–5766 (2010)CrossRefPubMed
50.
go back to reference H. Wehbe, C.M. Kearney, K.G. Pinney, Combretastatin A-4 resistance in H460 human lung carcinoma demonstrates distinctive alterations in β-tubulin isotype expression. Anticancer Res. 25, 3865–3870 (2005)PubMed H. Wehbe, C.M. Kearney, K.G. Pinney, Combretastatin A-4 resistance in H460 human lung carcinoma demonstrates distinctive alterations in β-tubulin isotype expression. Anticancer Res. 25, 3865–3870 (2005)PubMed
51.
go back to reference E.C. Tampaki, L. Nakopoulou, A. Tampakis, K. Kontzoglou, W.P. Weber, G. Kouraklis, Nestin involvement in tissue injury and cancer - a potential tumor marker? Cell. Oncol. 37, 305–315 (2014)CrossRef E.C. Tampaki, L. Nakopoulou, A. Tampakis, K. Kontzoglou, W.P. Weber, G. Kouraklis, Nestin involvement in tissue injury and cancer - a potential tumor marker? Cell. Oncol. 37, 305–315 (2014)CrossRef
52.
go back to reference D.O. Moon, M.O. Kim, C.H. Kang, J.D. Lee, Y.H. Choi, G.Y. Kim, JNK inhibitor SP600125 promotes the formation of polymerized tubulin, leading to G2/M phase arrest, endoreduplication, and delayed apoptosis. Exp. Mol. Med. 41, 665–677 (2009)PubMedCentralCrossRefPubMed D.O. Moon, M.O. Kim, C.H. Kang, J.D. Lee, Y.H. Choi, G.Y. Kim, JNK inhibitor SP600125 promotes the formation of polymerized tubulin, leading to G2/M phase arrest, endoreduplication, and delayed apoptosis. Exp. Mol. Med. 41, 665–677 (2009)PubMedCentralCrossRefPubMed
53.
go back to reference A.V. Singh, M. Bandi, N. Raje, P. Richardson, M.A. Palladino, D. Chauhan, K.C. Anderson, A novel vascular disrupting agent plinabulin triggers JNK-mediated apoptosis and inhibits angiogenesis in multiple myeloma cells. Blood 117, 5692–5700 (2011)PubMedCentralCrossRefPubMed A.V. Singh, M. Bandi, N. Raje, P. Richardson, M.A. Palladino, D. Chauhan, K.C. Anderson, A novel vascular disrupting agent plinabulin triggers JNK-mediated apoptosis and inhibits angiogenesis in multiple myeloma cells. Blood 117, 5692–5700 (2011)PubMedCentralCrossRefPubMed
54.
go back to reference L. Ciani, P.C. Salinas, c-Jun N-terminal kinase (JNK) cooperates with Gsk3β to regulate dishevelled-mediated microtubule stability. BMC Cell Biol. 8, 27 (2007)PubMedCentralCrossRefPubMed L. Ciani, P.C. Salinas, c-Jun N-terminal kinase (JNK) cooperates with Gsk3β to regulate dishevelled-mediated microtubule stability. BMC Cell Biol. 8, 27 (2007)PubMedCentralCrossRefPubMed
55.
go back to reference D.C.H. Ng, T.T. Zhao, Y.Y.C. Yeap, K.R. Ngoei, M.A. Bogoyevitch, c-Jun N-terminal kinase phosphorylation of stathmin confers protection against cellular stress. J. Biol. Chem. 285, 29001–29013 (2010)PubMedCentralCrossRefPubMed D.C.H. Ng, T.T. Zhao, Y.Y.C. Yeap, K.R. Ngoei, M.A. Bogoyevitch, c-Jun N-terminal kinase phosphorylation of stathmin confers protection against cellular stress. J. Biol. Chem. 285, 29001–29013 (2010)PubMedCentralCrossRefPubMed
56.
go back to reference Y.Y. Yip, Y.Y.C. Yeap, M.A. Bogoyevitch, D.C.H. Ng, Differences in c-Jun N-terminal kinase recognition and phosphorylation of closely related stathmin-family members. Biochem. Biophys. Res. Commun. 446, 248–254 (2014)CrossRefPubMed Y.Y. Yip, Y.Y.C. Yeap, M.A. Bogoyevitch, D.C.H. Ng, Differences in c-Jun N-terminal kinase recognition and phosphorylation of closely related stathmin-family members. Biochem. Biophys. Res. Commun. 446, 248–254 (2014)CrossRefPubMed
57.
go back to reference J.-H. Kim, T.H. Kim, H.S. Kang, J. Ro, H.S. Kim, S. Yoon, SP600125, an inhibitor of Jnk pathway, reduces viability of relatively resistant cancer cells to doxorubicin. Biochem. Biophys. Res. Commun. 387, 450–455 (2009)CrossRefPubMed J.-H. Kim, T.H. Kim, H.S. Kang, J. Ro, H.S. Kim, S. Yoon, SP600125, an inhibitor of Jnk pathway, reduces viability of relatively resistant cancer cells to doxorubicin. Biochem. Biophys. Res. Commun. 387, 450–455 (2009)CrossRefPubMed
58.
go back to reference X. Wang, K. Belguise, N. Kersual, K.H. Kirsch, N.D. Mineva, F. Galtier, D. Chalbos, G.E. Sonenshein, Oestrogen signalling inhibits invasive phenotype by repressing RelB and its target BCL2. Nat. Cell Biol. 9, 470–478 (2007)PubMedCentralCrossRefPubMed X. Wang, K. Belguise, N. Kersual, K.H. Kirsch, N.D. Mineva, F. Galtier, D. Chalbos, G.E. Sonenshein, Oestrogen signalling inhibits invasive phenotype by repressing RelB and its target BCL2. Nat. Cell Biol. 9, 470–478 (2007)PubMedCentralCrossRefPubMed
59.
go back to reference H. Lee, J. Jeon, Y.S. Ryu, J.E. Jeong, S. Shin, T. Zhang, S.W. Kang, J.H. Hong, G.M. Hur, Disruption of microtubules sensitizes the DNA damage-induced apoptosis through inhibiting nuclear factor kappaB (NF-κB) DNA-binding activity. J. Korean Med. Sci. 25, 1574 (2010)PubMedCentralCrossRefPubMed H. Lee, J. Jeon, Y.S. Ryu, J.E. Jeong, S. Shin, T. Zhang, S.W. Kang, J.H. Hong, G.M. Hur, Disruption of microtubules sensitizes the DNA damage-induced apoptosis through inhibiting nuclear factor kappaB (NF-κB) DNA-binding activity. J. Korean Med. Sci. 25, 1574 (2010)PubMedCentralCrossRefPubMed
60.
go back to reference S. Papa, Linking JNK signaling to NF-kappaB: a key to survival. J. Cell Sci. 117, 5197–5208 (2004)CrossRefPubMed S. Papa, Linking JNK signaling to NF-kappaB: a key to survival. J. Cell Sci. 117, 5197–5208 (2004)CrossRefPubMed
61.
go back to reference L. Li, B.B. Aggarwal, S. Shishodia, J. Abbruzzese, R. Kurzrock, Nuclear factor-kappaB and IkappaB kinase are constitutively active in human pancreatic cells, and their down-regulation by curcumin (diferuloylmethane) is associated with the suppression of proliferation and the induction of apoptosis. Cancer 101, 2351–2362 (2004)CrossRefPubMed L. Li, B.B. Aggarwal, S. Shishodia, J. Abbruzzese, R. Kurzrock, Nuclear factor-kappaB and IkappaB kinase are constitutively active in human pancreatic cells, and their down-regulation by curcumin (diferuloylmethane) is associated with the suppression of proliferation and the induction of apoptosis. Cancer 101, 2351–2362 (2004)CrossRefPubMed
Metadata
Title
Role of JNK and NF-κB in mediating the effect of combretastatin A-4 and brimamin on endothelial and carcinoma cells
Authors
Katharina Mahal
Aamir Ahmad
Seema Sethi
Marcus Resch
Ralf Ficner
Fazlul H. Sarkar
Rainer Schobert
Bernhard Biersack
Publication date
01-12-2015
Publisher
Springer Netherlands
Published in
Cellular Oncology / Issue 6/2015
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-015-0243-7

Other articles of this Issue 6/2015

Cellular Oncology 6/2015 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