Skip to main content

Advertisement

Log in

A novel structural derivative of natural alkaloid ellipticine, MDPSQ, induces necrosis in leukemic cells

  • PRECLINICAL STUDIES
  • Published:
Investigational New Drugs Aims and scope Submit manuscript

Summary

DNA intercalating molecules are promising chemotherapeutic agents. In the present study, a novel DNA intercalating compound of pyrimido[4′,5′:4,5]selenolo(2,3-b)quinoline series having 8-methyl-4-(3 diethylaminopropylamino) side chain is studied for its chemotherapeutic properties. Our results showed that 8-methyl-4-(3 diethylaminopropylamino) pyrimido [4′,5′:4,5] selenolo(2,3-b)quinoline (MDPSQ) induces cytotoxicity in a time- and concentration-dependent manner on leukemic cell lines. Both cell cycle analysis and tritiated thymidine assays revealed that MDPSQ affects DNA replication. Treatment with MDPSQ resulted in both elevated levels of DNA strand breaks and repair proteins, further indicating its cytotoxic effects. Besides, Annexin V/PI staining revealed that MDPSQ induces cell death by triggering necrosis rather than apoptosis.

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

Similar content being viewed by others

References

  1. Rowley JD (2001) Chromosome translocations: dangerous liaisons revisited. Nat Rev Cancer 1:245–250

    Article  PubMed  CAS  Google Scholar 

  2. Lieber MR (1993) In: Kirsch I (ed) The causes and consequences of chromosomal translocations. CRC, Boca Raton, pp 239–275

    Google Scholar 

  3. Raghavan SC, Lieber MR (2006) DNA structures at chromosomal translocation sites. Bioessays 28(5):480–494

    Article  PubMed  CAS  Google Scholar 

  4. Raghavan SC et al (2004) A non-B-DNA structure at the Bcl-2 major breakpoint region is cleaved by the RAG complex. Nature 428(6978):88–93

    Article  PubMed  CAS  Google Scholar 

  5. Nambiar M, Kari V, Raghavan SC (2008) Chromosomal translocations in cancer. Biochim Biophys Acta 1786(2):139–152

    PubMed  CAS  Google Scholar 

  6. Takeuchi Y et al (1997) Synthesis and antitumor activity of fused quinoline derivative. IV. Novel 11-aminoindolo(2, 3-b)quinolines. Chem Pharm Bull (Tokyo) 45:406–411

    CAS  Google Scholar 

  7. Auclair C et al (1987) Physicochemical and pharmacological properties of the antitumor ellipticine derivative 2-(diethylamino-2-ethyl)9-hydroxy ellipticinium-chloride. HCl. Cancer Res 47(23):6254–6261

    PubMed  CAS  Google Scholar 

  8. Tilak Raj T, Ambekar SY (1988) Synthesis of 4-amino pyrimido [4′, 5′:4, 5]thieno (2, 3-b) quinolines. J Chem Eng Data 33:530–531

    Article  Google Scholar 

  9. Tilak Raj T, Ambekar SY (1988) Synthesis of 4-amino pyrimido [4′, 5′:4, 5]thieno (2, 3-b) quinoline-4(3H)-ones. J Chem Res (S) 50:537–551

    Google Scholar 

  10. Dervan PB, Edelson BS (2003) Recognition of the DNA minor groove by pyrrole-imidazole polyamides. Curr Opin Struct Biol 13(3):284–299

    Article  PubMed  CAS  Google Scholar 

  11. Rezler EM, Bearss DJ, Hurley LH (2003) Telomere inhibition and telomere disruption as processes for drug targeting. Annu Rev Pharmacol Toxicol 43:359–379

    Article  PubMed  CAS  Google Scholar 

  12. Shahabuddin MS, Gopal M, Raghavan SC (2009) Intercalating, cytotoxic, antitumour activity of 8-chloro and 4-morpholinopyrimido [4′, 5′:4, 5]thieno(2, 3-b)quinolines. J Photochem Photobiol B 94(1):13–19

    Article  PubMed  CAS  Google Scholar 

  13. Shahabuddin MS et al (2009) A novel DNA intercalator, butylamino-pyrimido[4′,5′:4,5]selenolo(2,3-b)quinoline, induces cell cycle arrest and apoptosis in leukemic cells. Invest New Drugs

  14. Shahabuddin MS, Gopal M, Raghavan SC (2007) Intercalating and antitumor activity of 4-Oxopyrimido [4′, 5′:4, 5]thieno(2, 3-b)quinoline-4 (3H)-one. J Cancer Mol 3:139–146

    CAS  Google Scholar 

  15. Gopal M, Shahabuddin MS (2004) Biological properties of 8-methoxypyrimido[4(1), 5(1):4, 5]thieno(2, 3-b)quinoline-4(3H)-one, a new class of DNA intercalating drugs. Indian J Med Res 119(5):198–205

    PubMed  CAS  Google Scholar 

  16. Gopal M, Veeranna S (2005) 4-Anilinopyrimido[4′, 5′:4, 5]selenolo(2, 3-b)quinoline and 4-piperazino pyrimido[4′, 5′:4, 5]selenolo(2, 3-b)quinoline: new DNA intercalating chromophores with antiproliferative activity. J Photochem Photobiol B 81(3):181–189

    Article  PubMed  CAS  Google Scholar 

  17. Shenoy S et al (2007) 8-Methyl-4-(3-diethylaminopropylamino) pyrimido [4′, 5′;4, 5] thieno (2, 3-b) quinoline (MDPTQ), a quinoline derivate that causes ROS-mediated apoptosis in leukemia cell lines. Toxicol Appl Pharmacol 222(1):80–88

    Article  PubMed  CAS  Google Scholar 

  18. Gopal M, Shenoy S, Doddamani LS (2003) Antitumour activity of 4-amino and 8-methly-4-(3-diethyl aminopropylamino)pyrimido[4′, 5′:4, 5]thieno(2, 3-b)quinoline. J Photochem Photobiol B 72:69–78

    Article  PubMed  CAS  Google Scholar 

  19. Nandeeshaiah SK, Ambekar SY (1998) Synthesis, Dimroth rearrangment and blood platelet disaggregation property of pyrimido[4′, 5′:4, 5]selenolo(2, 3-b)quinolines: a new class of condensed quinoline. Indian J Chem 37:995–1000

    Google Scholar 

  20. Nandeeshaiah SK (1994) In: Department of Chemistry, University of Mysore, Mysore

  21. Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65(1–2):55–63

    Article  PubMed  CAS  Google Scholar 

  22. Chiruvella KK et al (2008) Methyl angolensate, a natural tetranortriterpenoid induces intrinsic apoptotic pathway in leukemic cells. FEBS Lett 582(29):4066–4076

    Article  PubMed  CAS  Google Scholar 

  23. Kavitha CV et al (2009) Novel derivatives of spirohydantoin induce growth inhibition followed by apoptosis in leukemia cells. Biochem Pharmacol 77(3):348–363

    Article  PubMed  CAS  Google Scholar 

  24. Vermes I et al (1995) A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J Immunol Methods 184(1):39–51

    Article  PubMed  CAS  Google Scholar 

  25. Mukhopadhyay P et al (2007) Simultaneous detection of apoptosis and mitochondrial superoxide production in live cells by flow cytometry and confocal microscopy. Nat Protoc 2(9):2295–2301

    Article  PubMed  CAS  Google Scholar 

  26. Kaufmann SH et al (1993) Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an early marker of chemotherapy-induced apoptosis. Cancer Res 53(17):3976–3985

    PubMed  CAS  Google Scholar 

  27. Ihmels H et al (2005) Intercalation of organic dye molecules into double-stranded DNA. Part 2: the annelated quinolizinium ion as a structural motif in DNA intercalators. Photochem Photobiol 81(5):1107–1115

    Article  PubMed  CAS  Google Scholar 

  28. Martinez R, Chacon-Garcia L (2005) The search of DNA-Intercalators as antitumoral drugs: what it worked and what did not work. Curr Med Chem 12(2):127–151

    PubMed  CAS  Google Scholar 

  29. Dervan PB et al (2005) Regulation of gene expression by synthetic DNA-binding ligands. Top Curr Chem 253:1–32

    CAS  Google Scholar 

  30. Fosse P et al (1992) Stimulation of topoisomerase II-mediated DNA cleavage by ellipticine derivatives: structure-activity relationship. Mol Pharmacol 42(4):590–595

    PubMed  CAS  Google Scholar 

  31. Juret P et al (1978) Preliminary trial of 9-hydroxy-2-methyl ellipticinium (NSC 264–137) in advanced human cancers. Eur J Cancer 14(2):205–206

    PubMed  CAS  Google Scholar 

  32. Mathe G et al (1970) Methoxy-9-ellipticine lactate. 3. Clinical screening: its action in acute myeloblastic leukaemia. Rev Eur Etud Clin Biol 15(5):541–545

    PubMed  CAS  Google Scholar 

  33. Brana MF et al (2001) Intercalators as anticancer drugs. Curr Pharm Des 7(17):1745–1780

    Article  PubMed  CAS  Google Scholar 

  34. Frei E et al (2002) Covalent binding of the anticancer drug ellipticine to DNA in V79 cells transfected with human cytochrome P450 enzymes. Biochem Pharmacol 64(2):289–295

    Article  PubMed  CAS  Google Scholar 

  35. Stiborova M et al (2001) The anticancer agent ellipticine on activation by cytochrome P450 forms covalent DNA adducts. Biochem Pharmacol 62(12):1675–1684

    Article  PubMed  CAS  Google Scholar 

  36. Ashkenazi A, Dixit VM (1998) Death receptors: signaling and modulation. Science 281(5381):1305–1308

    Article  PubMed  CAS  Google Scholar 

  37. Fantin VR, Leder P (2004) F16, a mitochondriotoxic compound, triggers apoptosis or necrosis depending on the genetic background of the target carcinoma cell. Cancer Res 64(1):329–336

    Article  PubMed  CAS  Google Scholar 

  38. Chiruvella KK et al (2007) Mechanism of DNA Double-Strand Break Repair. ICFAI J Biotech 1:7–22

    Google Scholar 

  39. Cepero V et al (2007) Trans-platinum(II) complexes with cyclohexylamine as expectator ligand induce necrosis in tumour cells by inhibiting DNA synthesis and RNA transcription. Clin Transl Oncol 9(8):521–530

    Article  PubMed  CAS  Google Scholar 

  40. Dartsch DC et al (2002) Comparison of anthracycline-induced death of human leukemia cells: programmed cell death versus necrosis. Apoptosis 7(6):537–548

    Article  PubMed  CAS  Google Scholar 

  41. Kiaris H, Schally AV (1999) Apoptosis versus necrosis: which should be the aim of cancer therapy? Proc Soc Exp Biol Med 221(2):87–88

    Article  PubMed  CAS  Google Scholar 

  42. Reiter I, Krammer B, Schwamberger G (1999) Cutting edge: differential effect of apoptotic versus necrotic tumor cells on macrophage antitumor activities. J Immunol 163(4):1730–1732

    PubMed  CAS  Google Scholar 

  43. Proskuryakov SY, Konoplyannikov AG, Gabai VL (2003) Necrosis: a specific form of programmed cell death? Exp Cell Res 283(1):1–16

    Article  PubMed  CAS  Google Scholar 

  44. Dewey WC, Ling CC, Meyn RE (1995) Radiation-induced apoptosis: relevance to radiotherapy. Int J Radiat Oncol Biol Phys 33(4):781–796

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Dr. Kavitha C.V., Ms. Nishana M. and members of SCR laboratory for critical reading of the manuscript. This work was supported by Lady Tata Memorial Trust international award for leukemia research (London) for SCR. SMS is supported by DBT postdoctoral fellowship from India. MN is supported by Senior Research Fellowship from CSIR, India.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sathees C. Raghavan.

Additional information

M. S. Shahabuddin and Mridula Nambiar contributed equally to the work.

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

Suppl. Figure 1

Quantification of cell cycle analysis of K562 and CEM cells following treatment with MDPSQ. K562 (A,B) and CEM (D,E) cells were treated with MDPSQ as described in Fig. 3. Quantification of different cell cycle phases is shown. C. Western blot showing expression of PCNA. The α-Tubulin was used as an internal loading control (PPT 283 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Shahabuddin, M.S., Nambiar, M., Moorthy, B.T. et al. A novel structural derivative of natural alkaloid ellipticine, MDPSQ, induces necrosis in leukemic cells. Invest New Drugs 29, 523–533 (2011). https://doi.org/10.1007/s10637-009-9379-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10637-009-9379-5

Keywords

Navigation