Skip to main content

Advertisement

Log in

Colchicine induces autophagy and senescence in lung cancer cells at clinically admissible concentration: potential use of colchicine in combination with autophagy inhibitor in cancer therapy

  • Original Article
  • Published:
Tumor Biology

Abstract

Colchicine is a well-known and potent microtubule targeting agent, but the therapeutic value of colchicine against cancer is limited by its toxicity against normal cells. But, there is no report of its cytotoxic potential against lung cancer cell, at clinically permissible or lower concentrations, minimally toxic to non-cancerous cells. Hence, in the present study, we investigated the possible mechanism by which the efficacy of colchicine against lung cancer cells at less toxic dose could be enhanced. Colchicine at clinically admissible concentration of 2.5 nM had no cytotoxic effect and caused no G2/M arrest in A549 cells. However, at this concentration, colchicine strongly hindered the reformation of cold depolymerised interphase and spindle microtubule. Colchicine induced senescence and reactive oxygen species mediated autophagy in A549 cells at this concentration. Autophagy inhibitor 3-methyladenine (3-MA) sensitised the cytotoxicity of colchicine in A549 cells by switching senescence to apoptotic death, and this combination had reduced cytotoxicity to normal lung fibroblast cells (WI38). Together, these findings indicated the possible use of colchicine at clinically relevant dose along with autophagy inhibitor in cancer therapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

References

  1. Cocco G, Chu DC, Pandolfi S. Colchicine in clinical medicine. A guide for internists. Eur J Int Med. 2010;21:503–8.

    Article  CAS  Google Scholar 

  2. Finkelstein Y, Aks SE, Hutson JR, Juurlink DN, Nguyen P, Dubnov-Raz G, et al. Colchicine poisoning: the dark side of an ancient drug. Clin Toxicol. 2010;48:407–14.

    Article  CAS  Google Scholar 

  3. Imazio M, Bobbio M, Cecchi E, Demarie D, Demichelis B, Pomari F, et al. Colchicine in addition to conventional therapy for acute pericarditis: results of the colchicine for acute pericarditis (cope) trial. Circulation. 2005;112:2012–6.

    Article  CAS  PubMed  Google Scholar 

  4. Kallinich T, Haffner D, Niehues T, Huss K, Lainka E, Neudorf U, et al. Colchicine use in children and adolescents with familial Mediterranean fever: literature review and consensus statement. Pediatrics. 2007;119:e474–83.

    Article  PubMed  Google Scholar 

  5. Yang LP. Oral colchicine (colcrys): in the treatment and prophylaxis of gout. Drugs. 2010;70:1603–13.

    Article  CAS  PubMed  Google Scholar 

  6. Roubille F, Kritikou E, Busseuil D, Barrere-Lemaire S, Tardif JC. Colchicine: an old wine in a new bottle? Anti-Inflamm Anti Allergy Agents Med Chem. 2013;12:14–23.

    Article  CAS  Google Scholar 

  7. Ravelli RB, Gigant B, Curmi PA, Jourdain I, Lachkar S, Sobel A, et al. Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature. 2004;428:198–202.

    Article  CAS  PubMed  Google Scholar 

  8. Bhattacharyya B, Panda D, Gupta S, Banerjee M. Anti-mitotic activity of colchicine and the structural basis for its interaction with tubulin. Med Res Rev. 2008;28:155–83.

    Article  CAS  PubMed  Google Scholar 

  9. Klement G, Baruchel S, Rak J, Man S, Clark K, Hicklin DJ, et al. Continuous low-dose therapy with vinblastine and vegf receptor-2 antibody induces sustained tumor regression without overt toxicity. J Clin Invest. 2000;105:R15–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gasparini G. Metronomic scheduling: the future of chemotherapy? Lancet Oncol. 2001;2:733–40.

    Article  CAS  PubMed  Google Scholar 

  11. Loven D, Hasnis E, Bertolini F, Shaked Y. Low-dose metronomic chemotherapy: from past experience to new paradigms in the treatment of cancer. Drug Discov Today. 2013;18:193–201.

    Article  CAS  PubMed  Google Scholar 

  12. Litwiniec A, Gackowska L, Helmin-Basa A, Zuryn A, Grzanka A. Low-dose etoposide-treatment induces endoreplication and cell death accompanied by cytoskeletal alterations in a549 cells: does the response involve senescence? the possible role of vimentin. Cancer Cell Int. 2013;13:9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sliwinska MA, Mosieniak G, Wolanin K, Babik A, Piwocka K, Magalska A, et al. Induction of senescence with doxorubicin leads to increased genomic instability of hct116 cells. Mech Ageing Dev. 2009;130:24–32.

    Article  CAS  PubMed  Google Scholar 

  14. Roninson IB. Tumor cell senescence in cancer treatment. Cancer Res. 2003;63:2705–15.

    CAS  PubMed  Google Scholar 

  15. Gewirtz DA, Holt SE, Elmore LW. Accelerated senescence: an emerging role in tumor cell response to chemotherapy and radiation. Biochem Pharmacol. 2008;76:947–57.

    Article  CAS  PubMed  Google Scholar 

  16. Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic ras and the p53 tumor suppressor. PLoS Biol. 2008;6:2853–68.

    Article  CAS  PubMed  Google Scholar 

  17. Elmore LW, Di X, Dumur C, Holt SE, Gewirtz DA. Evasion of a single-step, chemotherapy-induced senescence in breast cancer cells: implications for treatment response. Clin Cancer Res: Off J Am Assoc Cancer Res. 2005;11:2637–43.

    Article  CAS  Google Scholar 

  18. Roberson RS, Kussick SJ, Vallieres E, Chen SY, Wu DY. Escape from therapy-induced accelerated cellular senescence in p53-null lung cancer cells and in human lung cancers. Cancer Res. 2005;65:2795–803.

    Article  CAS  PubMed  Google Scholar 

  19. Seluanov A, Gorbunova V, Falcovitz A, Sigal A, Milyavsky M, Zurer I, et al. Change of the death pathway in senescent human fibroblasts in response to DNA damage is caused by an inability to stabilize p53. Mol Cell Biol. 2001;21:1552–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Abedin MJ, Wang D, McDonnell MA, Lehmann U, Kelekar A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2007;14:500–10.

    Article  CAS  PubMed  Google Scholar 

  21. Carew JS, Nawrocki ST, Kahue CN, Zhang H, Yang C, Chung L, et al. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor saha to overcome bcr-abl-mediated drug resistance. Blood. 2007;110:313–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, et al. Autophagy inhibition enhances therapy-induced apoptosis in a myc-induced model of lymphoma. J Clin Invest. 2007;117:326–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Akar U, Ozpolat B, Mehta K, Fok J, Kondo Y, Lopez-Berestein G. Tissue transglutaminase inhibits autophagy in pancreatic cancer cells. Mol Cancer Res MCR. 2007;5:241–9.

    Article  CAS  PubMed  Google Scholar 

  24. Moretti L, Attia A, Kim KW, Lu B. Crosstalk between bak/bax and mtor signaling regulates radiation-induced autophagy. Autophagy. 2007;3:142–4.

    Article  CAS  PubMed  Google Scholar 

  25. Gerland LM, Peyrol S, Lallemand C, Branche R, Magaud JP, Ffrench M. Association of increased autophagic inclusions labeled for beta-galactosidase with fibroblastic aging. Exp Gerontol. 2003;38:887–95.

    Article  CAS  PubMed  Google Scholar 

  26. Sasaki M, Miyakoshi M, Sato Y, Nakanuma Y. Autophagy mediates the process of cellular senescence characterizing bile duct damages in primary biliary cirrhosis. Lab Investig; J Tech Methods Pathol. 2010;90:835–43.

    Article  CAS  Google Scholar 

  27. Young AR, Narita M, Ferreira M, Kirschner K, Sadaie M, Darot JF, et al. Autophagy mediates the mitotic senescence transition. Genes Dev. 2009;23:798–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Arthur CR, Gupton JT, Kellogg GE, Yeudall WA, Cabot MC, Newsham IF, et al. Autophagic cell death, polyploidy and senescence induced in breast tumor cells by the substituted pyrrole jg-03-14, a novel microtubule poison. Biochem Pharmacol. 2007;74:981–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Alotaibi MR, Asnake B, Di X, Beckman MJ, Durrant D, Simoni D, et al. Stilbene 5c, a microtubule poison with vascular disrupting properties that induces multiple modes of growth arrest and cell death. Biochem Pharmacol. 2013;86:1688–98.

    Article  CAS  PubMed  Google Scholar 

  30. Sladowski D, Steer SJ, Clothier RH, Balls M. An improved mtt assay. J Immunol Methods. 1993;157:203–7.

    Article  CAS  PubMed  Google Scholar 

  31. Bhattacharya S, Kumar NM, Ganguli A, Tantak MP, Kumar D, Chakrabarti G. Nmk-td-100, a novel microtubule modulating agent, blocks mitosis and induces apoptosis in hela cells by binding to tubulin. PLoS One. 2013;8:e76286.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Banerjee M, Singh P, Panda D. Curcumin suppresses the dynamic instability of microtubules, activates the mitotic checkpoint and induces apoptosis in mcf-7 cells. FEBS J. 2010;277:3437–48.

    Article  CAS  PubMed  Google Scholar 

  33. Minotti AM, Barlow SB, Cabral F. Resistance to antimitotic drugs in Chinese hamster ovary cells correlates with changes in the level of polymerized tubulin. J Biol Chem. 1991;266:3987–94.

    CAS  PubMed  Google Scholar 

  34. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995;92:9363–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Munafo DB, Colombo MI. A novel assay to study autophagy: regulation of autophagosome vacuole size by amino acid deprivation. J Cell Sci. 2001;114:3619–29.

    CAS  PubMed  Google Scholar 

  36. Paglin S, Hollister T, Delohery T, Hackett N, McMahill M, Sphicas E, et al. A novel response of cancer cells to radiation involves autophagy and formation of acidic vesicles. Cancer Res. 2001;61:439–44.

    CAS  PubMed  Google Scholar 

  37. Karna P, Zughaier S, Pannu V, Simmons R, Narayan S, Aneja R. Induction of reactive oxygen species-mediated autophagy by a novel microtubule-modulating agent. J Biol Chem. 2010;285:18737–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kehrer JP, Paraidathathu T. The use of fluorescent probes to assess oxidative processes in isolated-perfused rat heart tissue. Free Radic Res Commun. 1992;16:217–25.

    Article  CAS  PubMed  Google Scholar 

  39. Picone R, Ren X, Ivanovitch KD, Clarke JD, McKendry RA, Baum B. A polarised population of dynamic microtubules mediates homeostatic length control in animal cells. PLoS Biol. 2010;8:e1000542.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Chang BD, Xuan Y, Broude EV, Zhu H, Schott B, Fang J, et al. Role of p53 and p21waf1/cip1 in senescence-like terminal proliferation arrest induced in human tumor cells by chemotherapeutic drugs. Oncogene. 1999;18:4808–18.

    Article  CAS  PubMed  Google Scholar 

  41. Mizushima N, Yoshimori T. How to interpret lc3 immunoblotting. Autophagy. 2007;3:542–5.

    Article  CAS  PubMed  Google Scholar 

  42. Scherz-Shouval R, Elazar Z. Ros, mitochondria and the regulation of autophagy. Trends Cell Biol. 2007;17:422–7.

    Article  CAS  PubMed  Google Scholar 

  43. Ferron GM, Rochdi M, Jusko WJ, Scherrmann JM. Oral absorption characteristics and pharmacokinetics of colchicine in healthy volunteers after single and multiple doses. J Clin Pharmacol. 1996;36:874–83.

    Article  CAS  PubMed  Google Scholar 

  44. Rochdi M, Sabouraud A, Girre C, Venet R, Scherrmann JM. Pharmacokinetics and absolute bioavailability of colchicine after i.V. And oral administration in healthy human volunteers and elderly subjects. Eur J Clin Pharmacol. 1994;46:351–4.

    Article  CAS  PubMed  Google Scholar 

  45. Terkeltaub RA, Furst DE, Bennett K, Kook KA, Crockett RS, Davis MW. High versus low dosing of oral colchicine for early acute gout flare: twenty-four-hour outcome of the first multicenter, randomized, double-blind, placebo-controlled, parallel-group, dose-comparison colchicine study. Arthritis Rheum. 2010;62:1060–8.

    Article  CAS  PubMed  Google Scholar 

  46. Lin ZY, Wu CC, Chuang YH, Chuang WL. Anti-cancer mechanisms of clinically acceptable colchicine concentrations on hepatocellular carcinoma. Life Sci. 2013;93:323–8.

    Article  CAS  PubMed  Google Scholar 

  47. Panda D, Roy S, Bhattacharyya B. Reversible dimer dissociation of tubulin s and tubulin detected by fluorescence anisotropy. Biochemistry. 1992;31:9709–16.

    Article  CAS  PubMed  Google Scholar 

  48. Le Grand M, Rovini A, Bourgarel-Rey V, Honore S, Bastonero S, Braguer D, et al. Ros-mediated eb1 phosphorylation through akt/gsk3beta pathway: implication in cancer cell response to microtubule-targeting agents. Oncotarget. 2014;5:3408–23.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

The work was supported by grants from DST, Govt. of India (No. SR/SO/BB-14/2008) and DBT, Government of India (No. BT/ PR12889/AGR/36/624/2009) to GC. Confocal Microscope facility is from University-DBT-IPLS programme, Government of India (No. BT/PR14552/INF/22/123/2010), and FACS instrument facility is developed by grants from National Common Minimum Project, Government of India. SB was a senior research fellow of University Grant Commission. SD is a Senior Research Fellow of CSIR, Government of India. AG was a fellow of DBT-CU-IPLS programmne, Government of India (No. BT/PR14552/INF/22/123/2010).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gopal Chakrabarti.

Ethics declarations

Conflicts of interest

None

Electronic supplementary material

Below is the link to the electronic supplementary material.

Fig. S1

Colchicine inhibited reformation of cold depolymerized interphase microtubule in A549 cells at low concentration Panel a shows normal interphase microtubule in A549 cells before incubating the cells at 4°C whereas panel b shows microtubule network after 3 h of cold depolymerization. Cold media was replaced by warm media in the absence (Panel c) and presence of 2.5 nM (Panel d) of colchicine and incubated for 3 h at 37°C. Microtubules were tagged by anti- α-tubulin antibody, subsequently FITC conjugated secondary antibody (green) and nucleus were probed by DAPI (blue). (GIF 42 kb)

High resolution image (TIF 1787 kb)

Fig. S2

Colchicine suppressed the reassembly of the cold depolymerized mitotic spindle microtubules in A549 cells Cultured A549 cells were incubated with nocodazole for 20h. After removing nocodazole, cells were kept on ice for 30 min in absence (Panel a-c) and presence of 2.5 nM colchicine (Panel g-i) and further incubated at 37°C in absence (Panel d-f) and presence of 2.5 nM colchicine (Panel j-l). (GIF 25 kb)

High resolution image (TIF 1533 kb)

Fig. S3

(a) Effect of colchicine on the ratio of soluble and polymerized tubulin in A549 cells. . A549 cells, treated without or with colchicine (2.5 and 50 nM), were lysed by a hypotonic lysis buffer. The insoluble and soluble tubulin fractions were separated by centrifugation. Western blot was performed using monoclonal antibody against α-tubulin. (b) Effect of colchicine on the ratio of soluble and polymerized tubulin in A549 cells before and after cold treatment. (c) Effect of 3-MA on the microtubule structure of A549 cells. Cultured A549 cells were incubated in absence and presence of 3-MA (5 mM) for 48 h, fixed and stained using antibody against α-tubulin (red) and nucleus with DAPI (blue). (GIF 15 kb)

High resolution image (TIF 1448 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bhattacharya, S., Das, A., Datta, S. et al. Colchicine induces autophagy and senescence in lung cancer cells at clinically admissible concentration: potential use of colchicine in combination with autophagy inhibitor in cancer therapy. Tumor Biol. 37, 10653–10664 (2016). https://doi.org/10.1007/s13277-016-4972-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-016-4972-7

Keywords

Navigation