Skip to main content

Advertisement

Log in

Targeting the HOX/PBX dimer in breast cancer

  • Preclinical Study
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

The HOX genes are a family of closely related transcription factors that help to define the identity of cells and tissues during embryonic development and which are also frequently deregulated in a number of malignancies, including breast cancer. While relatively little is known about the roles that individual HOX genes play in cancer, it is however clear that these roles can be both contradictory, with some members acting as oncogenes and some as tumor suppressors, and also redundant, with several genes essentially having the same function. Here, we have attempted to address this complexity using the HXR9 peptide to target the interaction between HOX proteins and PBX, a second transcription factor that serves as a common co-factor for many HOX proteins. We show that HXR9 causes apoptosis in a number of breast cancer-derived cell lines and that sensitivity to HXR9 is directly related to the averaged expression of HOX genes HOXB1 through to HOXB9, providing a potential biomarker to predict the sensitivity of breast tumors to HXR9 or its derivatives. Measuring the expression of HOX genes HOXB1HOXB9 in primary tumors revealed that a subset of tumors show highly elevated expression indicating that these might be potentially very sensitive to killing by HXR9. Furthermore, we show that while HXR9 blocks the oncogenic activity of HOX genes, it does not affect the known tumor-suppressor properties of a subset of HOX genes in breast cancer.

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. Abramovich C, Humphries RK (2005) Hox regulation of normal and leukemic hematopoietic stem cells. Curr Opin Hematol 12(3):210–216

    Article  PubMed  CAS  Google Scholar 

  2. Iimura T, Pourquie O (2007) Hox genes in time and space during vertebrate body formation. Dev Growth Differ 49(4):265–275

    Article  PubMed  CAS  Google Scholar 

  3. Moens CB, Selleri L (2006) Hox cofactors in vertebrate development. Dev Biol 291(2):193–206

    Article  PubMed  CAS  Google Scholar 

  4. Hoegg S, Meyer A (2005) Hox clusters as models for vertebrate genome evolution. Trends Genet 21(8):421–424

    Article  PubMed  CAS  Google Scholar 

  5. Scott MP (1993) A rational nomenclature for vertebrate homeobox (HOX) genes. Nucleic Acids Res 21(8):1687–1688

    Article  PubMed  CAS  Google Scholar 

  6. Shah N, Sukumar S (2010) The Hox genes and their roles in oncogenesis. Nat Rev Cancer 10(5):361–371

    Article  PubMed  CAS  Google Scholar 

  7. Chen H, Zhang H, Lee J, Liang X, Wu X, Zhu T, Lo PK, Zhang X, Sukumar S (2007) HOXA5 acts directly downstream of retinoic acid receptor beta and contributes to retinoic acid-induced apoptosis and growth inhibition. Cancer Res 67(17):8007–8013

    Article  PubMed  CAS  Google Scholar 

  8. Raman V, Martensen SA, Reisman D, Evron E, Odenwald WF, Jaffee E, Marks J, Sukumar S (2000) Compromised HOXA5 function can limit p53 expression in human breast tumours. Nature 405(6789):974–978

    Article  PubMed  CAS  Google Scholar 

  9. Care A, Silvani A, Meccia E, Mattia G, Peschle C, Colombo MP (1998) Transduction of the SkBr3 breast carcinoma cell line with the HOXB7 gene induces bFGF expression, increases cell proliferation and reduces growth factor dependence. Oncogene 16(25):3285–3289

    Article  PubMed  CAS  Google Scholar 

  10. Wu X, Chen H, Parker B, Rubin E, Zhu T, Lee JS, Argani P, Sukumar S (2006) HOXB7, a homeodomain protein, is overexpressed in breast cancer and confers epithelial–mesenchymal transition. Cancer Res 66(19):9527–9534

    Article  PubMed  CAS  Google Scholar 

  11. Jin K, Kong X, Shah T, Penet MF, Wildes F, Sgroi DC, Ma XJ, Huang Y, Kallioniemi A, Landberg G et al (2012) The HOXB7 protein renders breast cancer cells resistant to tamoxifen through activation of the EGFR pathway. Proc Natl Acad Sci USA 109(8):2736–2741

    Article  PubMed  CAS  Google Scholar 

  12. Galant R, Walsh CM, Carroll SB (2002) Hox repression of a target gene: extradenticle-independent, additive action through multiple monomer binding sites. Development 129(13):3115–3126

    PubMed  CAS  Google Scholar 

  13. Di-Poi N, Koch U, Radtke F, Duboule D (2010) Additive and global functions of HoxA cluster genes in mesoderm derivatives. Dev Biol 341(2):488–498

    Article  PubMed  CAS  Google Scholar 

  14. Lappin TR, Grier DG, Thompson A, Halliday HL (2006) HOX genes: seductive science, mysterious mechanisms. Ulster Med J 75(1):23–31

    PubMed  Google Scholar 

  15. Morgan R, Pirard PM, Shears L, Sohal J, Pettengell R, Pandha HS (2007) Antagonism of HOX/PBX dimer formation blocks the in vivo proliferation of melanoma. Cancer Res 67(12):5806–5813

    Article  PubMed  CAS  Google Scholar 

  16. Shears L, Plowright L, Harrington K, Pandha HS, Morgan R (2008) Disrupting the interaction between HOX and PBX causes necrotic and apoptotic cell death in the renal cancer lines CaKi-2 and 769-P. J Urol 180(5):2196–2201

    Article  PubMed  Google Scholar 

  17. Plowright L, Harrington KJ, Pandha HS, Morgan R (2009) HOX transcription factors are potential therapeutic targets in non-small-cell lung cancer (targeting HOX genes in lung cancer). Br J Cancer 100(3):470–475

    Article  PubMed  CAS  Google Scholar 

  18. Daniels TR, Neacato II, Rodriguez JA, Pandha HS, Morgan R, Penichet ML (2010) Disruption of HOX activity leads to cell death that can be enhanced by the interference of iron uptake in malignant B cells. Leukemia 24(9):1555–1565

    Article  PubMed  CAS  Google Scholar 

  19. Morgan R, Plowright L, Harrington KJ, Michael A, Pandha HS (2010) Targeting HOX and PBX transcription factors in ovarian cancer. BMC Cancer 10:89

    Article  PubMed  Google Scholar 

  20. Phelan ML, Sadoul R, Featherstone MS (1994) Functional differences between HOX proteins conferred by two residues in the homeodomain N-terminal arm. Mol Cell Biol 14(8):5066–5075

    PubMed  CAS  Google Scholar 

  21. Piper DE, Batchelor AH, Chang CP, Cleary ML, Wolberger C (1999) Structure of a HoxB1–Pbx1 heterodimer bound to DNA: role of the hexapeptide and a fourth homeodomain helix in complex formation. Cell 96(4):587–597

    Article  PubMed  CAS  Google Scholar 

  22. Knoepfler PS, Calvo KR, Chen H, Antonarakis SE, Kamps MP (1997) Meis1 and pKnox1 bind DNA cooperatively with Pbx1 utilizing an interaction surface disrupted in oncoprotein E2a–Pbx1. Proc Natl Acad Sci USA 94(26):14553–14558

    Article  PubMed  CAS  Google Scholar 

  23. Morgan R, In der Rieden P, Hooiveld MH, Durston AJ (2000) Identifying HOX paralog groups by the PBX-binding region. Trends Genet 16(2):66–67

    Article  PubMed  CAS  Google Scholar 

  24. Workman P, Balmain A, Hickman JA, McNally NJ, Rohas AM, Mitchison NA, Pierrepoint CG, Raymond R, Rowlatt C, Stephens TC et al (1988) UKCCCR guidelines for the welfare of animals in experimental neoplasia. Lab Animal 22(3):195–201

    Article  CAS  Google Scholar 

  25. White RA, Aspland SE, Brookman JJ, Clayton L, Sproat G (2000) The design and analysis of a homeotic response element. Mech Dev 91(1–2):217–226

    Article  PubMed  CAS  Google Scholar 

  26. Cailleau R, Young R, Olive M, Reeves WJ Jr (1974) Breast tumor cell lines from pleural effusions. J Natl Cancer Inst 53(3):661–674

    PubMed  CAS  Google Scholar 

  27. Cheng W, Liu J, Yoshida H, Rosen D, Naora H (2005) Lineage infidelity of epithelial ovarian cancers is controlled by HOX genes that specify regional identity in the reproductive tract. Nat Med 11(5):531–537

    Article  PubMed  CAS  Google Scholar 

  28. Gendronneau G, Lemieux M, Morneau M, Paradis J, Tetu B, Frenette N, Aubin J, Jeannotte L (2010) Influence of Hoxa5 on p53 tumorigenic outcome in mice. Am J Pathol 176(2):995–1005

    Article  PubMed  CAS  Google Scholar 

  29. Yokota I, Sasaki Y, Kashima L, Idogawa M, Tokino T (2010) Identification and characterization of early growth response 2, a zinc-finger transcription factor, as a p53-regulated proapoptotic gene. Int J Oncol 37(6):1407–1416

    PubMed  CAS  Google Scholar 

  30. Wang H, Mo P, Ren S, Yan C (2010) Activating transcription factor 3 activates p53 by preventing E6-associated protein from binding to E6. J Biol Chem 285(17):13201–13210

    Article  PubMed  CAS  Google Scholar 

  31. Suzuki M, Shigematsu H, Shames DS, Sunaga N, Takahashi T, Shivapurkar N, Iizasa T, Minna JD, Fujisawa T, Gazdar AF (2007) Methylation and gene silencing of the Ras-related GTPase gene in lung and breast cancers. Ann Surg Oncol 14(4):1397–1404

    Article  PubMed  Google Scholar 

  32. Zhang X, Pickin KA, Bose R, Jura N, Cole PA, Kuriyan J (2007) Inhibition of the EGF receptor by binding of MIG6 to an activating kinase domain interface. Nature 450(7170):741–744

    Article  PubMed  CAS  Google Scholar 

  33. Chen H, Lee JS, Liang X, Zhang H, Zhu T, Zhang Z, Taylor ME, Zahnow C, Feigenbaum L, Rein A et al (2008) Hoxb7 inhibits transgenic HER-2/neu-induced mouse mammary tumor onset but promotes progression and lung metastasis. Cancer Res 68(10):3637–3644

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to thank the Guy’s and St Thomas’ (GST) Breast Tissue and Data Bank, London, UK for providing the patient samples used in this study, and the patients who agreed to contribute to this data bank. The study was in part supported by a grant from the Breast Cancer Campaign to RM. The experiments carried out comply with the laws of the United Kingdom.

Conflict of interest

All authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard Morgan.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10549_2012_2259_MOESM1_ESM.eps

Online resource 1 Changes in expression of HXR9 target genes in SKBR3 cells. The relative expression of target genes are shown as a normalized intensity value (Log10), blue lines represent targets that increase expression in response to HXR9 compared to untreated cells [Unt], while the red lines represent targets that are repressed by HXR9 (EPS 142 kb)

10549_2012_2259_MOESM2_ESM.eps

Online resource 2 Distribution of HOX/PBX consensus sites in the promoter regions of the 20 most responsive HXR9 target genes. Possible HOX / PBX consensus binding sites are shown shaded according to the number of nucleotides that match the consensus (out of a maximum of 10). *Multiple consensus sites that overlap each other (EPS 205 kb)

10549_2012_2259_MOESM3_ESM.eps

Online resource 3 The mean expression values of HOX genes HOXB1 through to HOXB9 in primary tumors. Tumors were grouped according to (a) histopathological type, (b) estrogen receptor (ER) status, (c) spread to nodes as determined by pathology, (d) 5 year survival, (e) grade (G2, grade 2; G3, grade 3), (f) whether tumors were from the left or right breast, (g) progesterone receptor status (PR), and (h) HER2 status. Error bars show the SEM (EPS 260 kb)

Online resource 4–6 (DOC 101 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Morgan, R., Boxall, A., Harrington, K.J. et al. Targeting the HOX/PBX dimer in breast cancer. Breast Cancer Res Treat 136, 389–398 (2012). https://doi.org/10.1007/s10549-012-2259-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-012-2259-2

Keywords

Navigation