Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

A functional genomics approach to the mode of action of apratoxin A

Abstract

The cyanobacterial metabolite apratoxin A (1) demonstrates potent cytotoxicity against tumor cell lines by a hitherto unknown mechanism. We have used functional genomics to elucidate the molecular basis for this activity. Gene expression profiling and DNA content analysis showed that apratoxin A induces G1-phase cell cycle arrest and apoptosis. Cell-based functional assays with a genome-wide collection of expression cDNAs showed that ectopic induction of fibroblast growth factor receptor (FGFR) signaling attenuates the apoptotic activity of apratoxin A. This natural product inhibited phosphorylation and activation of STAT3, a downstream effector of FGFR signaling. It also caused defects in FGF-dependent processes during zebrafish development, with concomitant reductions in expression levels of the FGF target gene mkp3. We conclude that apratoxin A mediates its antiproliferative activity through the induction of G1 cell cycle arrest and an apoptotic cascade, which is at least partially initiated through antagonism of FGF signaling via STAT3.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Characterization of the biological effects of apratoxin A.
Figure 2: Genome-scale cDNA overexpression screen to identify proteins that confer resistance to apratoxin A.
Figure 3: Clusters of genes that are highly expressed in the cell lines most resistant to apratoxin A (OVCAR5, SK-MEL-2, OVCAR4) compared with four of the most susceptible cell lines (HT29, RPMI-8226, SR, LOX IMVI).
Figure 4: Effect of apratoxin A on STAT3 phosphorylation and transcriptional STAT3 activity in U2OS cells.
Figure 5: In vitro angiogenesis assay.
Figure 6: Effect of apratoxin A on zebrafish embryo development.

Similar content being viewed by others

References

  1. Newman, D.J., Cragg, G.M. & Snader, K.M. Natural products as sources of new drugs over the period 1981–2002. J. Nat. Prod. 66, 1022–1037 (2003).

    CAS  PubMed  Google Scholar 

  2. Jaspars, M. & Lawton, L.A. Cyanobacteria - a novel source of pharmaceuticals. Curr. Opin. Drug Discov. Devel. 1, 77–84 (1998).

    CAS  PubMed  Google Scholar 

  3. Liang, J. et al. Cryptophycins-309, 249 and other cryptophycin analogs: preclinical efficacy studies with mouse and human tumors. Invest. New Drugs 23, 213–224 (2005).

    CAS  PubMed  Google Scholar 

  4. Edelman, M.J. et al. Phase 2 study of cryptophycin 52 (LY355703) in patients previously treated with platinum based chemotherapy for advanced non-small cell lung cancer. Lung Cancer 39, 197–199 (2003).

    PubMed  Google Scholar 

  5. Luesch, H., Moore, R.E., Paul, V.J., Mooberry, S.L. & Corbett, T.H. Isolation of dolastatin 10 from the marine cyanobacterium Symploca species VP642 and total stereochemistry and biological evaluation of its analogue symplostatin 1. J. Nat. Prod. 64, 907–910 (2001).

    CAS  PubMed  Google Scholar 

  6. Vaishampayan, U. et al. Phase II study of dolastatin-10 in patients with hormone-refractory metastatic prostate adenocarcinoma. Clin. Cancer Res. 6, 4205–4208 (2000).

    CAS  PubMed  Google Scholar 

  7. Kavallaris, M., Verrills, N.M. & Hill, B.T. Anticancer therapy with novel tubulin-interacting drugs. Drug Resist. Updat. 4, 392–401 (2001).

    CAS  PubMed  Google Scholar 

  8. Luesch, H., Yoshida, W.Y., Moore, R.E., Paul, V.J. & Corbett, T.H. Total structure determination of apratoxin A, a potent novel cytotoxin from the marine cyanobacterium Lyngbya majuscula. J. Am. Chem. Soc. 123, 5418–5423 (2001).

    CAS  PubMed  Google Scholar 

  9. Luesch, H., Yoshida, W.Y., Moore, R.E. & Paul, V.J. New apratoxins of marine cyanobacterial origin from Guam and Palau. Bioorg. Med. Chem. 10, 1973–1978 (2002).

    CAS  PubMed  Google Scholar 

  10. Chen, J. & Forsyth, C.J. Total synthesis of the marine cyanobacterial cyclodepsipeptide apratoxin A. Proc. Natl. Acad. Sci. USA 101, 12067–12072 (2004).

    CAS  PubMed  Google Scholar 

  11. Paull, K.D., Hamel, E. & Malspeis, L. Prediction of biochemical mechanism of action from the in vitro antitumor screen of the National Cancer Institute. in Cancer Chemotherapeutic Agents (ed. Foye, W.O.) 9–45 (American Chemical Society Books, Washington, DC, 1995).

    Google Scholar 

  12. Beissbarth, T. & Speed, T.P. GOstat: find statistically overrepresented gene ontologies within a group of genes. Bioinformatics 20, 1464–1465 (2004).

    CAS  PubMed  Google Scholar 

  13. Lauper, N. et al. Cyclin E2: a novel CDK2 partner in the late G1 and S phases of the mammalian cell cycle. Oncogene 17, 2637–2643 (1998).

    CAS  PubMed  Google Scholar 

  14. Jinno, S. et al. Cdc25A is a novel phosphatase functioning early in the cell cycle. EMBO J. 13, 1549–1556 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Collavin, L., Monte, M., Verardo, R., Pfleger, C. & Schneider, C. Cell-cycle regulation of the p53-inducible gene B99. FEBS Lett. 481, 57–62 (2000).

    CAS  PubMed  Google Scholar 

  16. Lum, P.Y. et al. Discovering modes of action for therapeutic compounds using a genome-wide screen of yeast heterozygotes. Cell 116, 121–137 (2004).

    CAS  PubMed  Google Scholar 

  17. Giaever, G. et al. Chemogenomic profiling: identifying the functional interactions of small molecules in yeast. Proc. Natl. Acad. Sci. USA 101, 793–798 (2004).

    CAS  PubMed  Google Scholar 

  18. Luesch, H. et al. A genome-wide overexpression screen in yeast for small-molecule target identification. Chem. Biol. 12, 55–63 (2005).

    CAS  PubMed  Google Scholar 

  19. Choy, B.K., McClarty, G.A., Chan, A.K., Thelander, L. & Wright, J.A. Molecular mechanisms of drug resistance involving ribonucleotide reductase: hydroxyurea resistance in a series of clonally related mouse cell lines selected in the presence of increasing drug concentrations. Cancer Res. 48, 2029–2035 (1988).

    CAS  PubMed  Google Scholar 

  20. Espinet, C., Gómez-Arbonés, X., Egea, J. & Comella, J.X. Combined use of the green and yellow fluorescent proteins and fluorescence-activated cell sorting to select populations of transiently transfected PC12 cells. J. Neurosci. Methods 100, 63–69 (2000).

    CAS  PubMed  Google Scholar 

  21. Chambers, J.M. & Hastie, T.J. Statistical Models in S Ch. 8 (Chapman & Hall/CRC, London, 1992).

    Google Scholar 

  22. Eisen, M.B., Spellman, P.T., Brown, P.O. & Botstein, D. Cluster analysis and display of genome-wide expression patterns. Proc. Natl. Acad. Sci. USA 95, 14863–14868 (1998).

    CAS  Google Scholar 

  23. Hart, K.C. et al. Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene 19, 3309–3320 (2000).

    CAS  PubMed  Google Scholar 

  24. Yu, H. & Jove, R. The STATs of cancer – new molecular targets come of age. Nat. Rev. Cancer 4, 97–105 (2004).

    CAS  PubMed  Google Scholar 

  25. Zhong, Z., Wen, Z. & Darnell, J.E., Jr. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science 264, 95–98 (1994).

    CAS  PubMed  Google Scholar 

  26. Wen, Z., Zhong, Z. & Darnell, J.E., Jr. Maximal activation of transcription by Stat1 and Stat3 requires both tyrosine and serine phosphorylation. Cell 82, 241–250 (1995).

    CAS  PubMed  Google Scholar 

  27. Bromberg, J.F. et al. Stat3 as an oncogene. Cell 98, 295–303 (1999).

    CAS  PubMed  Google Scholar 

  28. Presta, M. et al. Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 16, 159–178 (2005).

    CAS  PubMed  Google Scholar 

  29. Xie, T. et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene 23, 3550–3560 (2004).

    CAS  PubMed  Google Scholar 

  30. Salani, D. et al. Endothelin-1 induces an angiogenic phenotype in cultured endothelial cells and stimulates neovascularization in vivo. Am. J. Pathol. 157, 1703–1711 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Dimitroff, C.J. et al. Anti-angiogenic activity of selected receptor tyrosine kinase inhibitors, PD166285 and PD173074: implications for combination treatment with photodynamic therapy. Invest. New Drugs 17, 121–135 (1999).

    CAS  PubMed  Google Scholar 

  32. Martin, G.R. The roles of FGFs in the early development of vertebrate limbs. Genes Dev. 12, 1571–1586 (1998).

    CAS  PubMed  Google Scholar 

  33. Capdevila, J. & Izpisúa Belmonte, J.C. Patterning mechanisms controlling vertebrate limb development. Annu. Rev. Cell Dev. Biol. 17, 87–132 (2001).

    CAS  PubMed  Google Scholar 

  34. Draper, B.W., Stock, D.W. & Kimmel, C.B. Zebrafish fgf24 functions with fgf8 to promote posterior mesodermal development. Development 130, 4639–4654 (2003).

    CAS  PubMed  Google Scholar 

  35. Reifers, F. et al. Fgf8 is mutated in zebrafish acerebellar (ace) mutants and is required for maintenance of midbrain-hindbrain boundary development and somitogenesis. Development 125, 2381–2395 (1998).

    CAS  PubMed  Google Scholar 

  36. Fischer, S., Draper, B.W. & Neumann, C.J. The zebrafish fgf24 mutant identifies an additional level of Fgf signaling involved in vertebrate forelimb initiation. Development 130, 3515–3524 (2003).

    CAS  PubMed  Google Scholar 

  37. Grandel, H., Draper, B.W. & Schulte-Merker, S. dackel acts in the ectoderm of the zebrafish pectoral fin bud to maintain AER signaling. Development 127, 4169–4178 (2000).

    CAS  PubMed  Google Scholar 

  38. Kawakami, Y. et al. MKP3 mediates the cellular response to FGF8 signalling in the vertebrate limb. Nat. Cell Biol. 5, 513–519 (2003).

    CAS  PubMed  Google Scholar 

  39. Tsang, M. et al. A role for MKP3 in axial patterning of the zebrafish embryo. Development 131, 2769–2779 (2004).

    CAS  PubMed  Google Scholar 

  40. Mohammadi, M. et al. Structures of the tyrosine kinase domain of fibroblast growth factor receptor in complex with inhibitors. Science 276, 955–960 (1997).

    CAS  PubMed  Google Scholar 

  41. Kawakami, Y. et al. Sp8 and Sp9, two closely related buttonhead-like transcription factors, regulate Fgf8 expression and limb outgrowth in vertebrate embryos. Development 131, 4763–4774 (2004).

    CAS  PubMed  Google Scholar 

  42. Tomono, M., Toyoshima, K., Ito, M., Amano, H. & Kiss, Z. Inhibitors of calcineurin block expression of cyclins A and E induced by fibroblast growth factor in Swiss 3T3 fibroblasts. Arch. Biochem. Biophys. 353, 374–378 (1998).

    CAS  PubMed  Google Scholar 

  43. Fuhrmann, G. et al. Cdc25A phosphatase suppresses apoptosis induced by serum deprivation. Oncogene 20, 4542–4553 (2001).

    CAS  PubMed  Google Scholar 

  44. Amin, H.M. et al. Selective inhibition of STAT3 induces apoptosis and G1 cell cycle arrest in ALK-positive anaplastic large cell lymphoma. Oncogene 23, 5426–5434 (2004).

    CAS  PubMed  Google Scholar 

  45. Catlett-Falcone, R. et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity 10, 105–115 (1999).

    CAS  PubMed  Google Scholar 

  46. Barré, B., Vigneron, A. & Coqueret, O. The STAT3 transcription factor is a target for the Myc and riboblastoma proteins on the Cdc25A promoter. J. Biol. Chem. 280, 15673–15681 (2005).

    PubMed  Google Scholar 

  47. Chan, K.S. et al. Disruption of Stat3 reveals a critical role in both the initiation and the promotion stages of epithelial carcinogenesis. J. Clin. Invest. 114, 720–728 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Xi, S., Gooding, W.E. & Grandis, J.R. In vivo antitumor efficacy of STAT3 blockade using a transcription factor decoy approach: implications for cancer therapy. Oncogene 24, 970–979 (2005).

    CAS  PubMed  Google Scholar 

  49. Westerfield, M. The Zebrafish Book: a Guide for the Laboratory Use of Zebrafish (Danio rerio) (Univ. of Oregon Press, Eugene, Oregon, 2000).

    Google Scholar 

  50. Hammerschmidt, M. et al. dino and mercedes, two genes regulating dorsal development in the zebrafish embryo. Development 123, 95–102 (1996).

    Google Scholar 

Download references

Acknowledgements

This work was supported by the Novartis Research Foundation (to P.G.S.), the US National Institutes of Health (NIH; to J.C.I.B.), the Irving S. Sigal Postdoctoral Fellowship (to H.L.), and an Inbiomed Fellowship (to R.M.R.). We would like to thank J. Zhang and S. Ho for the sample preparation for GeneChip analysis, A. Gutierrez for technical assistance in the logistics of the cDNA screen, S. White and A. Villar for providing reporter plasmids, P. McClurg for statistical analysis, C. Trussell for assisting in the FACS analysis, G. Xia for executing the FGFR kinase assay and for providing PD173074 and G. Hampton, E. Saez, T. Murphy and A. Willingham for helpful discussions. We would like to thank Y. Kawakami and Á. Raya for their support and helpful discussions regarding the zebrafish study, and C. Rodriguez for technical assistance in obtaining the zebrafish pictures. We also thank D. Newman for providing the NCI-60 data and for running the COMPARE analysis.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Hendrik Luesch or Peter G Schultz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Table 1

Activity of apratoxin A in the NCI-60 cytotoxicity assays. (PDF 33 kb)

Supplementary Table 2

Correlation of transcriptional changes induced in HT29 cells by various stress treatments and apratoxin A treatment (12 h). (PDF 60 kb)

Supplementary Table 3

cDNAs that attenuate the cytotoxicity of apratoxin A upon overexpression, grouped by their putative resistance mechanism. (PDF 31 kb)

Supplementary Table 4

Genes identified by hierarchical cluster analysis which are overexpressed in cancer cell lines most resistant to apratoxin A. (PDF 21 kb)

Supplementary Methods (PDF 89 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Luesch, H., Chanda, S., Raya, R. et al. A functional genomics approach to the mode of action of apratoxin A. Nat Chem Biol 2, 158–167 (2006). https://doi.org/10.1038/nchembio769

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio769

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing