Indibulin dampens microtubule dynamics and produces synergistic antiproliferative effect with vinblastine in MCF-7 cells: Implications in cancer chemotherapy

Sonia Kapoor, Shalini Srivastava, Dulal Panda, Sonia Kapoor, Shalini Srivastava, Dulal Panda

Abstract

Indibulin, a synthetic inhibitor of tubulin assembly, has shown promising anticancer activity with a minimal neurotoxicity in preclinical animal studies and in Phase I clinical trials for cancer chemotherapy. Using time-lapse confocal microscopy, we show that indibulin dampens the dynamic instability of individual microtubules in live breast cancer cells. Indibulin treatment also perturbed the localization of end-binding proteins at the growing microtubule ends in MCF-7 cells. Indibulin reduced inter-kinetochoric tension, produced aberrant spindles, activated mitotic checkpoint proteins Mad2 and BubR1, and induced mitotic arrest in MCF-7 cells. Indibulin-treated MCF-7 cells underwent apoptosis-mediated cell death. Further, the combination of indibulin with an anticancer drug vinblastine was found to exert synergistic cytotoxic effects on MCF-7 cells. Interestingly, indibulin displayed a stronger effect on the undifferentiated neuroblastoma (SH-SY5Y) cells than the differentiated neuronal cells. Unlike indibulin, vinblastine and colchicine produced similar depolymerizing effects on microtubules in both differentiated and undifferentiated SH-SY5Y cells. The data indicated a possibility that indibulin may reduce chemotherapy-induced peripheral neuropathy in cancer patients.

Trial registration: ClinicalTrials.gov NCT00591292 NCT00591136 NCT00591890 NCT00591383 NCT00726687 NCT01113970.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Indibulin inhibited the proliferation of MCF-7 cells and blocked the cell cycle progression at mitosis: (A) Structure of indibulin. (B) MCF-7 cells were treated with a vehicle or increasing concentrations of indibulin for 48 h. The inhibition of cell proliferation was determined by the SRB assay. Data are average of three independent experiments. (C) Cells were treated with vehicle or different concentrations (300 and 600 nM) of indibulin for 48 h and stained with propidium iodide. The DNA content of the cells was quantified by a flow cytometer and the data were analyzed using the Modfit LT program (Verity Software, ME, USA). The dark black lines show the fitting of the data by Modfit LT program and in each panel, the peaks correspond to G1 phase (left-side red peak), S phase (middle hashed line peak) and G2/M phase (right-side red peak). (D) Cells were treated with a vehicle or different concentrations of indibulin for 48 h and then stained with an antibody against phosphohistone H3 (S10) (green), a mitotic marker. DNA stained with Hoechst is shown in blue. Scale bar = 10 µm. (E) The histogram shows the percentage of phosphohistone H3 (S10) positive cells in the presence of a vehicle or different concentrations of indibulin. Three hundred cells were counted using Hoechst staining. Data are average of three independent experiments and error bar represents S.D.
Figure 2
Figure 2
Effects of indibulin on interphase microtubules in MCF-7 cells: (A) Cells were incubated with vehicle or different concentrations of indibulin for 24 h and were then stained with an antibody against α-tubulin (red). DNA stained with Hoechst is shown in blue. Scale bar = 10 µm. (B) MCF-7 cells were treated with vehicle or 150, 450 and 900 nM indibulin (lanes 1–4, respectively) for 48 h. 25 nM vinblastine (lane 5) was used under similar experimental conditions as a control. Polymeric and soluble tubulin fractions were isolated, loaded separately on two different SDS-PAGEs and immunoblotted with the α-tubulin antibody.
Figure 3
Figure 3
Effects of indibulin on mitotic MCF-7 cells: Cells were treated without and with different concentrations of indibulin and were stained with antibodies against α-tubulin (red) and γ-tubulin (green). Scale bar = 10 µm.
Figure 4
Figure 4
Indibulin suppressed microtubule dynamics in live MCF-7 cells: (A) Life history traces of microtubules of cells treated with (i) vehicle, (ii) 75 and (iii) 150 nM indibulin for 3 h. (B) Cells were treated with a vehicle or different concentrations (150 and 300 nM) of indibulin for 24 h and were processed for immunostaining with antibodies against EB1 (red) and α-tubulin (green). Boxed regions of merged panels are shown at higher magnification. Scale bar = 10 µm.
Figure 5
Figure 5
Indibulin activated the mitotic checkpoints in MCF-7 cells: (A,B) MCF-7 cells were incubated with a vehicle or different concentrations of indibulin for 24 h and processed for immunostaining with antibodies against Hec1 (red) and Mad2 (green) (A) or BubR1 (red) (B). Scale bar = 10 µm.
Figure 6
Figure 6
Indibulin increased the level of Mad2 and BubR1 and activated apoptosis in MCF-7 cells: (A) MCF-7 cells were incubated without and with different concentrations of indibulin and the cell extract was prepared. Vinblastine (25 nM) was used as a positive control. Western blot was performed with the extracts and immunoblotting was done with anti-Mad2 IgG, anti-BubR1 IgG, and anti-β actin IgG. The appropriate molecular weight bands of Mad2 (24 kDa), BubR1 (120 kDa) and β-actin (42 kDa) were cut from the same gel and immunoblotted with respective antibodies. The experiment was performed three times, shown is one of the representative blots. (B) MCF-7 cells were incubated with vehicle or different concentrations of indibulin for 48 h and then stained with annexin V (green) and propidium iodide (red) for detecting apoptosis. Scale bar = 10 µm. (C) Western blot of MCF-7 whole cell extracts prepared after incubating cells without and with different concentrations of indibulin and vinblastine (25 nM) for 48 h. The cell extract was separated on SDS-PAGE and appropriate molecular bands of PARP (116 kDa) and β-actin (42 kDa) were cut from the same gel. Immunoblotting was done with anti-PARP IgG and anti-β actin IgG. Shown is one of the representative blots from three experiments.
Figure 7
Figure 7
Indibulin and vinblastine exert synergistic anti-proliferative effects on MCF-7 cells. The median dose plot for the inhibition of cell proliferation in the presence of indibulin (i) and vinblastine (ii) is shown. Histogram (iii) shows the combination indices for the combination of indibulin (IN) with vinblastine (VB). Data are average of three independent experiments and represent mean ± SD.
Figure 8
Figure 8
Effects of indibulin on undifferentiated and differentiated SH-SY5Y cells. (A) Undifferentiated SH-SY5Y cells were incubated in the absence and presence of indibulin (25 nM), vinblastine (10 nM) and colchicine (10 nM) for 24 h. Cells were fixed and processed for immunostaining with antibodies against α-tubulin (green) and acetylated tubulin (red). Scale bar = 10 μm. (B) Differentiated SH-SY5Y cells were treated and processed as in (A) and observed at two magnifications under a microscope. Scale bar = 100 and 10 μm in low (Left panel) and high (Right panel) magnification, respectively.

References

    1. Bacher G, et al. D-24851, a novel synthetic microtubule inhibitor, exerts curative antitumoral activity in vivo, shows efficacy toward multidrug-resistant tumor cells, and lacks neurotoxicity. Cancer research. 2001;61:392–399.
    1. Ito H, Kanzawa T, Kondo S, Kondo Y. Microtubule inhibitor D-24851 induces p53-independent apoptotic cell death in malignant glioma cells through Bcl-2 phosphorylation and Bax translocation. International journal of oncology. 2005;26:589–596.
    1. Stokvis E, et al. Quantitative analysis of D-24851, a novel anticancer agent, in human plasma and urine by liquid chromatography coupled with tandem mass spectrometry. Rapid communications in mass spectrometry: RCM. 2004;18:1465–1471. doi: 10.1002/rcm.1493.
    1. Kuppens IE, et al. Phase I dose-finding and pharmacokinetic trial of orally administered indibulin (D-24851) to patients with solid tumors. Investigational new drugs. 2007;25:227–235. doi: 10.1007/s10637-006-9027-2.
    1. Oostendorp RL, et al. Dose-finding and pharmacokinetic study of orally administered indibulin (D-24851) to patients with advanced solid tumors. Investigational new drugs. 2010;28:163–170. doi: 10.1007/s10637-009-9244-6.
    1. Colley HE, et al. An Orally Bioavailable, Indole-3-glyoxylamide Based Series of Tubulin Polymerization Inhibitors Showing Tumor Growth Inhibition in a Mouse Xenograft Model of Head and Neck Cancer. Journal of medicinal chemistry. 2015;58:9309–9333. doi: 10.1021/acs.jmedchem.5b01312.
    1. Fanale D, et al. Stabilizing versus destabilizing the microtubules: a double-edge sword for an effective cancer treatment option? Analytical cellular pathology. 2015;2015:690916. doi: 10.1155/2015/690916.
    1. Huang TH, et al. Antiproliferative effects of N-heterocyclic indolyl glyoxylamide derivatives on human lung cancer cells. Anticancer research. 2011;31:3407–3415.
    1. Li WT, et al. Synthesis and biological evaluation of N-heterocyclic indolyl glyoxylamides as orally active anticancer agents. Journal of medicinal chemistry. 2003;46:1706–1715. doi: 10.1021/jm020471r.
    1. Kamath K, Oroudjev E, Jordan MA. Determination of microtubule dynamic instability in living cells. Methods in cell biology. 2010;97:1–14. doi: 10.1016/S0091-679X(10)97001-5.
    1. Kapoor S, Panda D. Kinetic stabilization of microtubule dynamics by indanocine perturbs EB1 localization, induces defects in cell polarity and inhibits migration of MDA-MB-231 cells. Biochemical pharmacology. 2012;83:1495–1506. doi: 10.1016/j.bcp.2012.02.012.
    1. Mohan R, Panda D. Kinetic stabilization of microtubule dynamics by estramustine is associated with tubulin acetylation, spindle abnormalities, and mitotic arrest. Cancer research. 2008;68:6181–6189. doi: 10.1158/0008-5472.CAN-08-0584.
    1. Kamath K, Jordan MA. Suppression of microtubule dynamics by epothilone B is associated with mitotic arrest. Cancer research. 2003;63:6026–6031.
    1. Rathinasamy K, Panda D. Suppression of microtubule dynamics by benomyl decreases tension across kinetochore pairs and induces apoptosis in cancer cells. The FEBS journal. 2006;273:4114–4128. doi: 10.1111/j.1742-4658.2006.05413.x.
    1. Walker RA, et al. Dynamic instability of individual microtubules analyzed by video light microscopy: rate constants and transition frequencies. The Journal of cell biology. 1988;107:1437–1448. doi: 10.1083/jcb.107.4.1437.
    1. Slep KC. Structural and mechanistic insights into microtubule end-binding proteins. Current opinion in cell biology. 2010;22:88–95. doi: 10.1016/j.ceb.2009.10.009.
    1. Zanic M, Stear JH, Hyman AA, Howard J. EB1 recognizes the nucleotide state of tubulin in the microtubule lattice. PloS one. 2009;4:e7585. doi: 10.1371/journal.pone.0007585.
    1. Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time. Nature reviews. Molecular cell biology. 2007;8:379–393. doi: 10.1038/nrm2163.
    1. Taylor SS, Hussein D, Wang Y, Elderkin S, Morrow CJ. Kinetochore localisation and phosphorylation of the mitotic checkpoint components Bub1 and BubR1 are differentially regulated by spindle events in human cells. Journal of cell science. 2001;114:4385–4395.
    1. Venghateri JB, Gupta TK, Verma PJ, Kunwar A, Panda D. Ansamitocin P3 depolymerizes microtubules and induces apoptosis by binding to tubulin at the vinblastine site. PloS one. 2013;8:e75182. doi: 10.1371/journal.pone.0075182.
    1. Gajula PK, Asthana J, Panda D, Chakraborty TK. A synthetic dolastatin 10 analogue suppresses microtubule dynamics, inhibits cell proliferation, and induces apoptotic cell death. Journal of medicinal chemistry. 2013;56:2235–2245. doi: 10.1021/jm3009629.
    1. Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG. Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer research. 1993;53:3976–3985.
    1. Kolomeichuk SN, Terrano DT, Lyle CS, Sabapathy K, Chambers TC. Distinct signaling pathways of microtubule inhibitors–vinblastine and Taxol induce JNK-dependent cell death but through AP-1-dependent and AP-1-independent mechanisms, respectively. The FEBS journal. 2008;275:1889–1899. doi: 10.1111/j.1742-4658.2008.06349.x.
    1. Wienecke A, Bacher G. Indibulin, a novel microtubule inhibitor, discriminates between mature neuronal and nonneuronal tubulin. Cancer research. 2009;69:171–177. doi: 10.1158/0008-5472.CAN-08-1342.
    1. Cheung YT, et al. Effects of all-trans-retinoic acid on human SH-SY5Y neuroblastoma as in vitro model in neurotoxicity research. Neurotoxicology. 2009;30:127–135. doi: 10.1016/j.neuro.2008.11.001.
    1. Presgraves SP, Ahmed T, Borwege S, Joyce JN. Terminally differentiated SH-SY5Y cells provide a model system for studying neuroprotective effects of dopamine agonists. Neurotoxicity research. 2004;5:579–598. doi: 10.1007/BF03033178.
    1. Jordan MA, Kamath K. How do microtubule-targeted drugs work? An overview. Current cancer drug targets. 2007;7:730–742. doi: 10.2174/156800907783220417.
    1. Schiff D, Wen PY, van den Bent MJ. Neurological adverse effects caused by cytotoxic and targeted therapies. Nature reviews. Clinical oncology. 2009;6:596–603. doi: 10.1038/nrclinonc.2009.128.
    1. Carlson K, Ocean AJ. Peripheral neuropathy with microtubule-targeting agents: occurrence and management approach. Clinical breast cancer. 2011;11:73–81. doi: 10.1016/j.clbc.2011.03.006.
    1. Hansen SW, Helweg-Larsen S, Trojaborg W. Long-term neurotoxicity in patients treated with cisplatin, vinblastine, and bleomycin for metastatic germ cell cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 1989;7:1457–1461. doi: 10.1200/JCO.1989.7.10.1457.
    1. Verstappen CC, et al. Dose-related vincristine-induced peripheral neuropathy with unexpected off-therapy worsening. Neurology. 2005;64:1076–1077. doi: 10.1212/01.WNL.0000154642.45474.28.
    1. Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (second of two parts) The New England journal of medicine. 1974;291:127–133. doi: 10.1056/NEJM197407182910305.
    1. Rathinasamy K, Panda D. Kinetic stabilization of microtubule dynamic instability by benomyl increases the nuclear transport of p53. Biochemical pharmacology. 2008;76:1669–1680. doi: 10.1016/j.bcp.2008.09.001.
    1. Skehan P, et al. New colorimetric cytotoxicity assay for anticancer-drug screening. Journal of the National Cancer Institute. 1990;82:1107–1112. doi: 10.1093/jnci/82.13.1107.
    1. Asthana J, Kapoor S, Mohan R, Panda D. Inhibition of HDAC6 deacetylase activity increases its binding with microtubules and suppresses microtubule dynamic instability in MCF-7 cells. The Journal of biological chemistry. 2013;288:22516–22526. doi: 10.1074/jbc.M113.489328.
    1. Rieger AM, Nelson KL, Konowalchuk JD, Barreda DR. Modified annexin V/propidium iodide apoptosis assay for accurate assessment of cell death. Journal of visualized experiments: JoVE. 2011
    1. Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. Journal of immunological methods. 1995;184:39–51. doi: 10.1016/0022-1759(95)00072-I.
    1. Harrill JA, Freudenrich TM, Machacek DW, Stice SL, Mundy WR. Quantitative assessment of neurite outgrowth in human embryonic stem cell-derived hN2 cells using automated high-content image analysis. Neurotoxicology. 2010;31:277–290. doi: 10.1016/j.neuro.2010.02.003.
    1. Ochoa CD, Stevens T, Balczon R. Cold exposure reveals two populations of microtubules in pulmonary endothelia. American journal of physiology. Lung cellular and molecular physiology. 2011;300:L132–138. doi: 10.1152/ajplung.00185.2010.
    1. Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Advances in enzyme regulation. 1984;22:27–55. doi: 10.1016/0065-2571(84)90007-4.
    1. Clement MJ, et al. Benomyl and colchicine synergistically inhibit cell proliferation and mitosis: evidence of distinct binding sites for these agents in tubulin. Biochemistry. 2008;47:13016–13025. doi: 10.1021/bi801136q.

Source: PubMed

3
Předplatit